METHYLENE CHLORIDE r c Agency for Toxic Substances and Disease Registry U.S. Public Health Service ------- ATSDR/TP-88/18 TOXICOLOGICAL PROFILE FOR METHYLENE CHLORIDE Date Published — April 1989 Prepared by: Life Systems, Inc. under Contract No. 68-02-4228 for Agency for Toxic Substances and Disease Registry (ATSDR) U.S. Public Health Service in collaboration with U.S. Environmental Protection Agency (EPA) Technical editing/document preparation by: Oak Ridge National Laboratory under DOE Interagency Agreement No. I857-B026-AI ------- DISCLAIMER Mention of company name or product does not constitute endorsement by the Agency for Toxic Substances and Disease Registry. ------- FOREWORD The Superfund Amendments and Reauthorizacion Act of 1986 (Public Law 99-499) extended and amended the Comprehensive Environmental Response, Compensation, and Liability Act of 1980 (CERCLA or Superfund) This public law (also known as SARA) directed the Agency for Toxic Substances and Disease Registry (ATSDR) to prepare toxicological profiles for hazardous substances which are most commonly found at facilities on the CERCLA National Priorities List and which pose the most significant potential threat to human health, as determined by ATSDR and the Environmental Protection Agency (EPA). The list of the 100 most significant hazardous substances was published in the Federal RegLscer on April 17, 1987. Section 110 (3) of SARA directs the Administrator of ATSDR to prepare a toxicological profile for each substance on the list. Each profile must include the following content: "(A) An examination, summary, and interpretation of available toxicological information and epidemiologic evaluations on a hazardous substance in order to ascertain the levels of significant human exposure for the substance and the associated acute, subacute, and chronic health effects. (B) A determination of whether adequate information on the health effects of each substance is available or in the process of development to determine levels of exposure which present a significant risk to human health of acute, subacute, and chronic health effects. (C) Where appropriate, an identification of toxicological testing needed to identify the types or levels of exposure that may present significant risk of adverse health effects in humans." This toxicological profile is prepared in accordance with guidelines developed by ATSDR and EPA. The guidelines were published in the Federal Register on April 17, 1987. Each profile will be revised and republished as necessary, but no less often than every three years, as required by SARA. The ATSDR toxicological profile is intended to characterize succinctly the toxicological and health effects information for the hazardous substance being described. Each profile identifies and reviews the key literature that describes a hazardous substance's toxicological properties. Other literature is presented but described in less detail than the key studies. The profile is not intended to be an exhaustive document; however, more comprehensive sources of specialty information are referenced. ILL ------- Foreword Each toxlcological profile begins with a public health statement, which describes in nontechnical language a substance's relevant toxicological properties. Following the statement is material that presents levels of significant human exposure and, where known, significant health effects. The adequacy of information to determine a substance's health effects is described in a health effects summary. Research gaps in toxicologic and health effects information are described in the profile. Research gaps that are of significance to protection of public health will be identified by ATSDR, the National Toxicology Program of the Public Health Service, and EPA. The focus of the profiles is on health and toxicological information; therefore, we have included this information in the front of the document. The principal audiences for the toxicological profiles are health professionals at the federal, state, and local levels, interested private sector organizations and groups, and members of the public. We plan to revise these documents in response to public comments and as additional data become available; therefore, we encourage comment that will make the toxicological profile series of the greatest use. This profile reflects our assessment of all relevant toxicological testing and information that has been peer reviewed. It has been reviewed by scientists from ATSDR, EPA, the Centers for Disease Control, and the National Toxicology Program. It has also been reviewed by a panel of nongovernment peer reviewers and was made available for public review. Final responsibility for the contents and views expressed in this toxicological profile resides with ATSDR. James 0. Mason, M.D., Dr. P.H. Assistant Surgeon General Administrator, ATSDR iv ------- CONTENTS FOREWORD iii LIST OF FIGURES '. . . ix LIST OF TABLES xi 1. PUBLIC HEALTH STATEMENT 1 1.1 WHAT IS METHYLENE CHLORIDE? 1 1.2 HOW MIGHT I BE EXPOSED TO METHYLENE CHLORIDE? 1 1.3 HOW DOES METHYLENE CHLORIDE GET INTO MY BODY? 1 1.4 HOW CAN METHYLENE CHLORIDE AFFECT MY HEALTH? 1 1.5 IS THERE A MEDICAL TEST TO DETERMINE IF I HAVE BEEN EXPOSED TO METHYLENE CHLORIDE? 2 1.6 WHAT LEVELS OF EXPOSURE HAVE RESULTED IN HARMFUL HEALTH EFFECTS , 2 1.7 WHAT RECOMMENDATIONS HAS THE FEDERAL GOVERNMENT MADE TO PROTECT HUMAN HEALTH? 5 2. HEALTH EFFECTS SUMMARY 7 2.1 INTRODUCTION 7 2.2 LEVELS OF SIGNIFICANT HUMAN EXPOSURE 8 2.2.1 Key Studies 8 2.2.1.1 Lethality 16 2.2.1.2 Systemic/target organ toxicity 16 2.2.1.3 Developmental toxicity 18 2.2.1.4 Genotoxicity 18 2.2.1.5 Reproductive toxicity 18 2.2.1.6 Carcinogenicity 19 2.2.1.7 Mechanisms of action 20 2.2.2 Biological Monitoring as a Measure of Exposure and Effects 21 2.2.3 Environmental Levels as Indicators of Exposure and Effects 21 2.2.3.1 Levels found in the environment 21 2.2.3.2 Human exposure potential 22 2.3 ADEQUACY OF DATABASE 22 2.3.1 Introduction 22 2.3.2 Health Effect End Points 23 2.3.2.1 Introduction and graphic summary 23 2.3.2.2 Description of highlights of graphs 23 2.3.2.3 Summary of relevant ongoing research .... 23 2.3.3 Other Information Needed for Human Health Assessment 26 2.3.3.1 Pharmacokinetics and mechanisms of action 26 2.3.3.2 Monitoring of human biological samples .. 26 2.3.3.3 Environmental considerations 26 ------- Concents 3. PHYSICAL AND CHEMICAL INFORMATION 27 3 .1 CHEMICAL IDENTITY ' ' 27 3 . 2 PHYSICAL AND CHEMICAL PROPERTIES '.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'. 27 4. TOXICOLOGICAL DATA 31 4.1 OVERVIEW 31 4. 2 TOXICOKINETICS '.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'. 32 4.2.1 Overview 32 4.2.2 Absorption 33 4.2.2.1 Inhalation '.'.'.'.'.'. 33 4.2.2.2 Oral 34 4.2.2.3 Dermal 35 4.2.3 Distribution " 35 4.2.3.1 Inhalation 35 4.2.3.2 Oral '.'.'.'.'.'.'.'.'. 36 4.2.3.3 Dermal 35 4.2.4 Metabolism '.'.'.'.'.'.'.'.'.'.'.'. 36 4.2.4.1 Inhalation 36 4.2.4.2 Oral '.'.'.'.'.'.'.'.'. 42 4.2.4.3 Dermal 43 4.2.4.4 In vitro studies 44 4.2.5 Excretion 44 4.2.5.1 Inhalation 44 4.2.5.2 Oral '.'.'.'.'.'.'.'.'.'. 46 4.2.5.3 Dermal 46 4.2.6 Discussion 45 4. 3 TOXICITY '.'.'.'.'.'.'.'.'.'.'.'.'. 47 4.3.1 Overview 47 4.3.2 Lethality and Decreased Longevity 47 4.3.2.1 Overview 47 4.3.2.2 Inhalation 47 4.3.2.3 Oral "'"' 43 4.3.2.4 Dermal 48 4.3.2.5 Discussion 48 4.3.3 Systemic/Target Organ Toxicity 48 4.3.3.1 Overview 43 4.3.3.2 Central nervous system 50 4.3.3.3 Hepatotoxicity 53 4.3.3.4 Renal effects 58 4.3.3.5 Respiratory effects 59 4.3.3.6 Cardiovascular 59 4.3.3.7 Ocular effects 60 4.3.4 Developmental Toxicity 60 4.3.4.1 Overview 60 4.3.4.2 Inhalation 60 4.3.4.3 Oral 61 4.3.4.4 Dermal 61 4.3.4.5 Discussion 61 4.3.5 Reproductive Toxicity 61 4.3.5.1 Inhalation 61 4.3.5.2 Oral 62 4.3.5.3 Dermal 62 4.3.5.4 Discussion 62 4.3.6 Genotoxicity 62 vi ------- Concents 4.3.7 Carcinogenicity 65 4.3.7.1 Overview 65 4.3.7.2 Human 65 4.3.7.3 Animal 66 4.4 INTERACTIONS WITH OTHER CHEMICALS 68 5. MANUFACTURE, IMPORT, USE, AND DISPOSAL 69 5.1 OVERVIEW 69 5.2 PRODUCTION 69 5.3 IMPORT 69 5.4 USE 69 5.5 DISPOSAL 70 6. ENVIRONMENTAL FATE 71 6 .1 OVERVIEW " 71 6 . 2 RELEASES TO THE ENVIRONMENT 71 6.2.1 Anthropogenic Sources 71 6.2.2 Natural Sources 72 6.3 ENVIRONMENTAL FATE 72 6.3.1 Atmospheric Fate Processes 72 6.3.2 Surface Water/Groundwater Fate Processes 72 6.3.3 Soil Fate Processes 73 6.3.4 Biotic Fate Processes 73 7. POTENTIAL FOR HUMAN EXPOSURE 75 7.1 OVERVIEW 75 7.2 LEVELS MONITORED OR ESTIMATED IN THE ENVIRONMENT 75 7.2.1 Levels in Air 75 7.2.2 Levels in Water 76 7.2.3 Levels in Soil 76 7.2.4 Levels in Food 76 7.2.5 Resulting Exposure Levels 76 7.3 OCCUPATIONAL EXPOSURES 77 7.4 CONSUMER EXPOSURE 77 7. 5 POPULATIONS AT HIGH RISK 80 7.5.1 Above-Average Exposure 80 7.5.2 Above-Average Sensitivity 80 8. ANALYTICAL METHODS 81 8.1 ENVIRONMENTAL MEDIA 81 8.1.1 Air 81 8.1.2 Water 83 8.1.3 Soil 83 8.1.4 Food 83 8.2 BIOMEDICAL SAMPLES 83 8.2.1 Fluids and Exudates 83 8.2.2 Tissues 83 9. REGULATORY AND ADVISORY STATUS 85 9.1 INTERNATIONAL 85 9.2 NATIONAL 85 9.2.1 Regulations 85 9.2.2 Advisory Guidance 88 9.2.3 Data Analysis 89 9.2.3.1 References doses 89 9.2.3.2 Carcinogenic potency 90 vll ------- Concents 9.3 STATE 9Q 9.3.1 Regulations 90 9.3.2 Advisory Guidance 90 10. REFERENCES 91 11. GLOSSARY 107 APPENDIX: PEER REVIEW m viii ------- LIST OF FIGURES 1.1 Health effects from breathing methylene chloride 3 1.2 Health effects from ingesting methylene chloride 4 2.1 Effects of methylene chloride--inhalation exposure 12 2.2 Levels of significant exposure for methylene chloride-- inhalation 13 2.3 Effects of methylene chloride--oral exposure 14 2.4 Levels of significant exposure for methylene chloride-- oral 15 2.5 Availability of information on health effects of methylene chloride (human data) 24 2.6 Availability of information on health effects of methylene chloride (animal data) 25 4.1 Proposed pathways for methylene chloride metabolism 45 ------- LIST OF TABLES 2 1 Key dose response data for short- terra CNS effects of methylene chloride ................................... 1-0 2.2 Key dose response data for long-term hepatotoxicity effects of methylene chloride ............................... 11 3 1 Chemical identity of methylene chloride .................... 28 3 2 Physical and chemical properties of methylene chloride ...... 29 4.1 Comparison of average daily values of dose surrogates in lung and liver tissue of female B6C3F1 mice in two chronic bioassays ....................................... 41 4 . 2 Acute lethality of methylene chloride ....................... 49 4.3 Short- terra experimental methylene chloride inhalation exposures and reported effects in humans ..................... 51 4.4 Longer-term occupational methylene chloride inhalation exposures and effects in humans .............................. 52 4.5 Short-term methylene chloride inhalation exposures and effects in animals ...................................... 54 4.6 Longer-term methylene chloride inhalation exposures and effects in animals ....................................... 57 4.7 Results of methylene chloride testing in various mammalian short-term tests .................................. 63 4.8 Summary of primary tumors in rats and mice in 2 -year studies of methylene chloride ......................... 67 7.1 Occupations in which methylene chloride exposures may occur [[[ 78 7.2 Summary of occupational exposure to methylene chloride ....... 79 8.1 Analytical methods for methylene chloride in environmental samples [[[ 82 8.2 Analytical methods for methylene chloride in biological ------- 1. PUBLIC HEALTH STATEMENT 1.1 WHAT IS METHYLENE CHLORIDE? Methylene chloride, also known as dichloromethane (DCM), is an organic solvent that looks like water, has a mild sweet odor, and evaporates very quickly. It is widely used as an industrial solvent and as a paint stripper. It is also a component in certain aerosol and pesticide products and is used in the manufacture of photographic film. 1.2 HOW MIGHT I BE EXPOSED TO METHYLENE CHLORIDE? The highest exposures to DCM usually occur in workplaces where DCM is used or from contact with consumer products that contain DCM; exposure to this solvent in outdoor air and water is generally low. Hobby and household use of paint-stripping chemicals and DCM-containing aerosol products are major sources of exposure. Exposure occurs as a result of breathing the vapors given off by the product or from direct contact of the product material with the skin. Special efforts should be taken to follow label directions that recommend working in a well- ventilated area when using products containing DCM. Also, some decaffeinated coffee and spices contain small amounts of DCM; however, exposures from these sources are considered insignificant. 1.3 HOW DOES METHYLENE CHLORIDE GET INTO MY BODY? Methylene chloride may enter the body when it is inhaled or ingested. No data are currently available on the absorption of DCM through human skin. Since DCM vaporizes very quickly, the primary route of exposure is through inhalation. Once DCM enters the body, it is absorbed through body membranes (e.g., stomach, intestines, and lungs) and quickly enters the bloodstream. 1.4 HOW CAN METHYLENE CHLORIDE AFFECT MY HEALTH? High levels of DCM in air (above about 500 ppm) can irritate the eyes, nose, and throat. If DCM gets on the skin, it usually evaporates quickly and causes only mild irritation. However, DCM can be trapped against the skin by gloves, shoes, or clothes and can cause a burn. If DCM gets into the eyes, it may cause a severe (but temporary) eye irritation. Methylene chloride can affect the central nervous system (brain). If DCM is breathed at levels above about 500 ppm (500 parts DCM per 1 million parts air), it may cause effects much like those produced by alcohol, including sluggishness, irritability, lightheadedness, nausea. and headaches. Some effects have been observed at concentrations as low as 300 ppm. These symptoms usually disappear quite rapidly after exposure ends. ------- 2 Section I Some of the effects to the nervous system caused by DCM may be because of the breakdown of DCM In the body to carbon monoxide (CO) Carbon monoxide Interferes with the blood's ability to carry oxygen (02) to the tissues and causes symptoms similar to the narcotic effects previously described. Since smoking Increases the amount of CO in the blood, smokers may experience effects to the nervous system at lower levels of DCM exposure than do nonsmokers. The results of animal studies suggest that frequent or lengthy exposures to DCM can cause changes in the liver and kidney. However, based on these studies and those of exposed workers, it appears unlikely that DCM will cause serious liver or kidney damage in humans unless exposure is very high. No liver or kidney effects vere reported in humans exposed to 30 to 125 ppm DCM in the workplace for up to 30 years. or to 140 to 475 ppm for at least 3 months. In animal studies, mild liver effects were observed from 100 ppm (100 days) to 3,500 ppm (2 years). It should be noted that hepatic effects reported at 100 ppm were elicited after continuous exposure without recovery as opposed to the more traditional exposure protocol. In certain laboratory experiments, animals exposed to high concentrations of DCM throughout their lifetime developed cancer. Methylene chloride has not been shown to cause cancer in humans exposed at occupational levels; however, based on animal tests, it should be treated as a potential cancer-causing substance. 1.5 IS THERE A MEDICAL TEST TO DETERMINE IF I HAVE BEEN EXPOSED TO METHYLENE CHLORIDE? Several methods exist for determining whether a person has recently been exposed to DCM. Methylene chloride can be measured In the breath co determine recent exposure; the amount of chemical detected will reflect the amount inhaled. The urine can also be analyzed by monitoring for DCM itself or for some intermediate products (such as formic acid) that are produced as DCM breaks down in the body. Blood can be analyzed to determine possible DCM exposure by monitoring blood levels of carboxyhemoglobin (CO-Hb). Carbon monoxide formed in the blood through the breakdown of DCM readily binds with hemoglobin to form CO-Hb. Thus, excessive levels of CO-Hb in the blood can be an indication of exposure to high concentrations of DCM. Although exposure to DCM can be monitored through these sources, measurements determined have not provided definitive quantitative information. 1.6 WHAT LEVELS OF EXPOSURE HAVE RESULTED IN HARMFUL HEALTH EFFECTS? The graphs on the following pages (Figs. 1.1 and 1.2) show the relationship between exposure to DCM and known health effects. In the first set of graphs labeled "Health effects from breathing methylene chloride," exposure is measured In parts of chemical per million pares of air (ppm). In all graphs, effects in animals are shown on the left side and effects In humans on the right side. ------- Public Health Statement SHORT-TERM EXPOSURE (LESS THAN OR EQUAL TO 14 DAYS) LONG-TERM EXPOSURE (GREATER THAN 14 DAYS) EFFECTS IN ANIMALS CONC IN AIR (ppm) 100.000 EFFECTS IN HUMANS EFFECTS IN ANIMALS CONC IN AIR (ppm) 100.000 DEATH CNS EFFECTS • EFFECTS IN HUMANS QUANTITATIVE DATA WERE NOT AVAILABLE ON EXPOSURE TO DCM ALONE 10.000 10.000 LIVER TOXICITY • CNS EFFECTS 1.000 1.000 •CNS EFFECTS •CNS EFFECTS 100 LIVER TOXICITY • 100' 10 10 'Continuous exposure Fig. 1.1. Health effects from breathing methylene chloride. ------- Seed on 1 SHORT-TERM EXPOSURE (LESS THAN OR EQUAL TO 14 DAYS) LONG-TERM EXPOSURE (GREATER THAN 14 DAYS) EFFECTS IN ANIMALS DOSE (mg/kg/day) QUANTITATIVE DATA WERE NOT AVAILABLE 1000 EFFECTS IN HUMANS QUANTITATIVE DATA WERE NOT AVAILABLE EFFECTS IN ANIMALS DOSE (mg/kg/day) 1000 100 LIVER TOXICITY 100 10 10 1 0 1 0 EFFECTS IN HUMANS QUANTITATIVE DATA WERE NOT AVAILABLE Fig. 1.2. Health effects from ingesting methylene chloride. ------- Public Health Statement 5 In the second set of graphs, the same relationship is represented for the known "Health effects from eating or drinking products containing methylene chloride." Exposures are measured in milligrams of DCM per kilogram of body weight (mg/kg). In case studies involving humans, the primary health effects are on the central nervous system. Short exposures to concentrations of SOO ppm (duration not specified) and above result in chemical intoxication, tiredness, and irritability. One study reported a slight effect on sensory function at 300 ppm (4 hours). At concentrations of 500 ppm (duration not specified) and above, DCM also irritates the nose and throat. Rats have developed changes in liver cells following long-term ingestion of 50 mg/kg/day DCM in drinking water. However, based on animal and human studies, it appears unlikely that DCM will cause serious liver effects in humans unless exposure is very high. Methylene chloride is not known to cause cancer in humans, but based on animal studies, the Environmental Protection Agency (EPA) believes that DCM has the potential to cause cancer in humans. Exposures should, therefore, be avoided or, when unavoidable, kept to the lowest level feasible. 1.7 WHAT RECOMMENDATIONS HAS THE FEDERAL GOVERNMENT MADE TO PROTECT HUMAN HEALTH? The Occupational Health and Safety Administration (OSHA 1979) has established exposure limits for persons who work with DCM. These include an 8-hour time-weighted average (TWA) of 1,737 milligrams per cubic meter (mg/m3) (500 ppm); an acceptable ceiling concentration of 3,474 mg/m3 (1,000 ppm); and an acceptable maximum peak above the ceiling of 6,948 mg/m3 (2,000 ppm) (5 minutes in any 2 hours) in the workplace air OSHA is currently considering revising these standards based on the recent information available on cancer studies in animals. In 1976, the National Institute for Occupational Safety and Health (NIOSH 1976) recommended a 10-hour TWA exposure limit of 261 mg/m3 (75 ppm) apd a 1,737 mg/m3 (500 ppm) peak (15-minute sampling) in the presence of CO concentrations less than or equal to 9.9 ppm. These recommendations were made because DCM and CO are responsible for the same kinds of toxic effects. Lower levels of DCM are required in the workplace when CO concentrations greater than 9.9 ppm are present. Because DCM has been shown to induce increased numbers of benign and malignant neoplasms in rats and mice, NIOSH currently recommends that the compound be considered a potential human carcinogen in the workplace and that the TWA exposure limit of 75 ppm be reduced to the lowest feasible limit. ------- 2. HEALTH EFFECTS SUMMARY 2.1 INTRODUCTION This section summarizes and graphs data on the health effects concerning exposure to DCM. The purpose of this section is to present levels of significant exposure for DCM based on key toxicological studies, epidemiological investigations, and environmental exposure data. The information presented in this section is critically evaluated and discussed in Sect. 4, Toxicological Data, and Sect. 7, Potential for Human Exposure. This Health Effects Summary section comprises two major parts. Levels of Significant Exposure (Sect. 2.2) presents brief narratives and graphics for key studies in a manner that provides public health officials, physicians, and other interested individuals and groups wich (1) an overall perspective of the toxicology of DCM and (2) a summarized depiction of significant exposure levels associated with various adverse health effects. This section also includes information on the levels of DCM that have been monitored in human fluids and tissues and information about levels of DCM found in environmental media and their association with human exposures. The significance of the exposure levels shown on the graphs may differ depending on the user's perspective. For example, physicians concerned with the interpretation of overt clinical findings in exposed persons or with the identification of persons with the potential to develop such disease may be interested in levels of exposure associated with frank effects (Frank Effect Level, FEL). Public health officials and project managers concerned with response actions at Superfund sites may want information on levels of exposure associated with more subtle effects in humans or animals (Lowest-Observed-Adverse-Effect Level, LOAEL) or exposure levels below which no adverse effects (No-Observed- Adverse-Effect Level, NOAEL) have been observed. Estimates of levels posing minimal risk to humans (Minimal Risk Levels) are of interest to health professionals and citizens alike. Adequacy of Database (Sect. 2.3) highlights the availability of key studies on exposure to DCM in the scientific literature and displays these data in three-dimensional graphs consistent with the format in Sect. 2.2. The purpose of this section is to suggest where there might be insufficient information to establish levels of significant human exposure. These areas will be considered by the Agency for Toxic Substances and Disease Registry (ATSDR), EPA, and the National Toxicology Program (NTP) of the U.S. Public Health Service in order to develop a research agenda for DCM. ------- 8 Section 2 2.2 LEVELS OF SIGNIFICANT HUNAN EXPOSURE To help public health professionals address the needs of persons living or working near hazardous waste sites, the toxicology data summarized in this section are organized first by route of exposure-- inhalation, ingestion, and dermal--and then by toxicological end points that are categorized into six general areas--lethality, systemic/target organ toxicity, developmental toxicity, reproductive toxicity, genetic toxicity, and carcinogenicity. The data are discussed in terms of three exposure periods--acute, intermediate, and chronic. Two kinds of graphs are used to depict the data. The first type is a "thermometer" graph. It provides a graphical summary of the human and animal toxicological end points (and levels of exposure) for each exposure route for which data are available. The ordering of effects does not reflect the exposure duration or species of animal tested. The second kind of graph shows Levels of Significant Exposure (LSE) for each route and exposure duration. The points on the graph showing NOAELs and LOAELs reflect the actual doses (levels of exposure) used in the key studies. No adjustments for exposure duration or intermittent exposure protocol were made. Adjustments reflecting the uncertainty of extrapolating animal data to man, intraspecies variations, and differences between experimental vs actual human exposure conditions were considered when estimates of levels posing minimal risk to human health were made for noncancer end points. These minimal risk levels were derived for the most sensitive noncancer end point for each exposure duration by applying uncertainty factors. These levels are shown on the graphs as a broken line starting from the actual dose (level of exposure) and ending with a concave- curved line at its terminus. Although methods have been established to derive these minimal risk levels (Barnes et al. 1987), shortcomings exist in the techniques that reduce the confidence in the projected estimates. Also shown on the graphs under the cancer end point are low- levels risks (10'4 to 10'7) reported by EPA. In addition, the actual dose (level of exposure) associated with the tumor incidence is plotted. 2.2.1 Key Studies Humans exposed to high concentrations of DCM for short time periods may experience effects on the central nervous system. In case reports of humans exposed to DCM through inhalation, the major effects noted were narcosis, irritability, analgesia, and fatigue. Short-term exposure to about 300 to 800 ppm resulted in impairment of the sensory and psychomotor function (Winneke 1974). Data on laboratory animals also indicate that DCM has a "depressive" effect on the central nervous system. The lowest concentration reported to cause effects was 1,000 ppm (Fodor and Winneke 1971). At this dose, the sleep patterns of rats were affected. The no-effect level in this study was established as 500 ppm. Acute inhalation exposures to high concentrations (4,000 to 20,000 ppm) of DCM have produced symptoms of central nervous system depression (including narcosis) in several other species, including mice (Flury and Zernik 1931), guinea pigs (Weinstein et al. 1972), and dogs (Weinstein ec al. 1972, Von Oettingen et al. ------- Health Effects Summary 9 Liver effects caused by exposure Co DCM have been documented in repeated exposure studies on laboratory animals. Mild cytoplasmic vacuolization and/or fatty changes were observed in the liver of mice, rats, dogs, and monkeys following inhalation exposure. These effects were seen following exposure of rats and dogs to 25 to 100 ppm for 100 days Twenty-five ppm was a no-effect level for mice; 100 ppm was a no- effect level for monkeys (Haun et al. 1972). Foci, or areas of altered morphology, fatty change, or necrosis, were observed in the liver of rats and mice following long-term exposure (2-year inhalation study). Lowest-observed-effect levels ranged from 500 to 1,000 ppm 6 h/day 5 days/week in rats (Nitschke et al. 1982, NTP 1986). Five mg/kg/day was an oral no-effect level in rats, and 185 mg/kg/day was an oral no-effect level in mice (NCA 1982, 1983). Nonspecific degenerative and regenerative changes were observed in the kidneys of rats exposed to 25 or 100 ppm continuously for 100 days (Haun et al. 1972). Exposure to high concentrations of DCM can cause carboxyhemoglobinemia, a condition resulting from carbon monoxide (CO) interference with oxygen transport in the blood. Metabolism of DCM produces CO, which readily binds with blood hemoglobin to form carboxyhemoglobin (CO-Hb). The National Institute for Occupational Safety and Health (NIOSH) has recommended that exposure to DCM should not produce CO-Hb levels that exceed 5% (i.e., no more than 5% of the blood's hemoglobin should be saturated with CO). A study (Ott et al. 1983c) on workers exposed to DCM through their jobs in a fiber-extrusion plant provided information on the relationship between DCM exposure and CO-Hb buildup, both in smokers and nonsmokers. Hemoglobin saturation following exposure to 200 ppm DCM was about 4% in nonsmokers and 10% in smokers. In those exposed to 100 ppm, saturation was 2% in nonsmokers and 6% in smokers. At 50 ppm, saturation was slightly more than 4% in smokers but less than 1% in nonsmokers. Data from this study indicate that exposures to DCM concentrations below 50 ppm are not likely to result in the buildup of CO-Hb above the NIOSH- recommended limit of 5%, either in smokers or nonsmokers. Recently conducted long-term studies clearly demonstrate that DCM causes cancer in laboratory animals (NTP 1986). Mice exposed via inhalation to high concentrations of DCM (2,000 or 4,000 ppm) experienced a dramatic increase in liver and lung malignant tumors compared with mice that were not exposed to DCM. Rats exposed via inhalation to DCM (1,000, 2,000, or 4,000 ppm) developed mammary gland tumors, but these tumors were benign. Methylene chloride as a cancer- causing agent in humans has not been established. Studies of workers exposed to DCM for several years have not recorded a significant increase in cancer cases above the number of cases expected for nonexposed workers. However, these occupational studies are not adequate to rule out a risk of cancer to humans from DCM exposure, and this chemical should be regarded as a possible carcinogen in humans. The primary adverse health effects associated with exposure to DCM are depression of the central nervous system and mild liver toxicity. These effects are summarized in Tables 2.1 and 2.2, and data from these studies are presented graphically in Figs. 2.1 through 2.4. ------- 10 Section 2 Table 2.1. Key dose response data for short-term CNS effects of methylene chloride Route of exposure Species Inhalation Human Inhalation Human Rat Dose-duration LOAEL" - (4h) NOAEL* = LOAEL = LOAEL = NOAEL =- (24 h) 300-800 ppm 1 500 ppm 1,000 ppm 1,000-3,000 ppm 500 ppm Effect Impairment of visual, auditory, and psychomotor functions Decreased visual function Reduction in sleeping time References Winneke 1974 Stewart et al. 1972 Fodor and Winneke 1971 Oral Dermal No data available on humans and animals No data available on humans and animals • LOAEL •" lowest-observed-adverse-effect level. * NOAEL = no-observed-adverse-effect level. ------- Healch Effects Summary 11 Table 2.2. Key dote response data for long-term bepatotoxkity effects of metbylene chloride Route of exposure Species Dose-response Effect References Inhalation Inhalation Inhalation Inhalation Mouse Monkey Rat Dog Rat Rat Rat NOAEL - 25 ppm LOAEL - 100 ppm (100 days) NOAEL - 100 ppm LOAEL - 1,000 ppm (100 days) LOAEL - 25 ppm (100 days) NOAEL - 200 ppm LOAEL - 500 ppm (2 years) LOAEL - 1. 000 ppm (2 years) LOAEL - 500-3.500 ppm (2 years) Histopathological changes in liver. including vacuoli- zation and increased fat content Weight reduction in mice. Multmucleated hepatocytes Increased hemosiderosu* cytomegaly. and cytoplasm ic vacuoluation Multmucleated hepatocytes with cytoplasmic vacuolization. hemosiderosis, and focal necrosis Haun et al 1972 Nitschke et al. 1982 NTP 1986 Burek et al. 1980, 1984 Oral (drinking water) Oral (drinking water) Oral Denial Human Rat Mouse Human Cases of occupational exposure were presented. but dose-response relationships were not defined NOAEL - 5 rag/kg/day LOAEL - 50-250 mg/kg/day (2 yean) NOAEL - 185 mg/kg/day LOAEL - 250 mg/kg/day Histomorpnological alterations in liver Fatty changes in the liver No data available No data available on humans or animals NCA 1982 NCA 1983 ------- 12 Section 2 ANIMALS 10.000 r— • MICE CNS EFFECTS • GUINEA PIGS CNS EFFECTS • DOGS CNS EFFECTS 1000 100 10 *- HUMANS (PP"i) 10000 i— f RATS. CNS EFFECTS. 24 h - •< RATS. UVER EFFECTS. 2 YEARS I MONKEYS. UVER EFFECTS. 100 DAYS (• RATS LIVER EFFECTS. 2 YEARS IJi RATS. CNS EFFECTS. 100 DAYS ' MICE LIVER TOXICITY. 100 DAYS • RATS. KIDNEY EFFECTS. 100 DAYS > MONKEYS. LIVER EFFECTS. 100 DAYS !• RATS. KIDNEY EFFECTS. 100 DAYS J« RATS. DOGS LIVER EFFECTS, 100 DAYS (p MICE LIVER TOXICITY. 100 DAYS 1000 100 • LOAEL FOR ANIMALS O NOAEL FOR ANIMALS A LOAEL FOR HUMANS A NOAEL FOR HUMANS 10 •— A CNS EFFECTS 5 h £ UVER LUNG (>20 YEARS) 'Continuous exposure Fig. 2.1. Effects of methylene chloride—inhalation exposure. ------- Health Effects Summary 13 (ppm) 10.000 1 000 100 10 1 0 01 001 0001 - 00001 - 0 00001 «- ACUTE (<14 DAYS) TARGET ORGAN • m (CMS) • g(CNS) •d (CMS) •r (CNS) INTERMEDIATE (15-364 DAYS) CHRONIC (>365 DAYS) TARGET ORGAN REPRO- DUCTION TARGET ORGAN CANCER f (CNS) m(LIVER) I k (LIVER) I r (LIVER) • r (KIDNEY. LIVER) d (LIVER) 10-5- 10-6- ESTIMATED UPPER-BOUND HUMAN CANCER RISK LEVELS r RAT m MOUSE k MONKEY d DOG g GUINEA PIG A LOAEL FOR HUMANS • LOAEL FOR ANIMALS O NOAEL FOR ANIMALS I I MINIMAL RISK LEVEL FOR I EFFECTS OTHER THAN \llCANCER Fig. 2.2. Levels of significant exponrc for nettaylene chloride—inhaladoo. ------- 14 Seccion 2 ANIMALS (mg/kg/day) 1000 i— HUMANS 100 • MOUSE. LIVER EFFECTS. 2 YEARS O MOUSE LIVER EFFECTS. 2 YEARS RAT. LIVER EFFECTS. 2 YEARS 10 O RAT. LIVER EFFECTS. 2 YEARS 10 L- • LOAEL ONOAEL QUANTITATIVE DATA WERE NOT AVAILABLE Fig. 2J. Effects of methylene chloride—oral exposure. ------- Health Effects Siunmary 15 (mg/kg/day) LOOOr- 100 10 0.1 0.01 0.001 00001 0.00001 !- ACUTE INTERMEDIATE (S14 DAYS) (15-364 DAYS) LETHALITY • r m MOUSE r RAT QUANTITATIVE DATA WERE NOT AVAILABLE • LOAEL O NOAEL CHRONIC (>365 DAYS) TARGET ORGAN • m (LIVER) O r (LIVER) CANCER • m 10-4-, 10-5- 10 -7- ESTIMATED UPPER-BOUND HUMAN CANCER RISK LEVELS MINIMAL RISK LEVEL FOR EFFECTS OTHER THAN CANCER Fig. 2.4. Levcb of significant exposure for methykM chloride—onL ------- 16 Section 2 2.2.1.L Lethality Inhalation, human. Case reports (Bonventre et al. 1977, Stewart and Hake 1976) have implicated exposure to exceedingly high levels of DCM as a factor in human fatalities. However, exposure levels and durations are not known, and simultaneous exposure to unspecified chemicals was often the case. Inhalation, animal. Studies demonstrate that DCM is lethal in laboratory animals via inhalation. Inhalation LC50 values of 11,000 to 16,000 ppm (for 6 to 8 h) have been reported (EPA 1985c) (Table 4.2). Oral, human. No lethality studies were found in the available literature on the oral administration of DCM in humans. Oral, animal. Oral LD50 values of 1,000 to 2,000 mg/kg have been reported (EPA 1985c) (Table 4.1). Oral, dermal. No lethality studies were found in the available literature on the dermal administration of DCM in humans and animals. 2.2.1.2 Systemic/target organ toxicity Central nervous system, inhalation (human). The major manifestation in humans of short-term exposure to DCM is the impairment in the functioning of the central nervous system. Winneke (1974) used indicators of behavioral performance to study the effects of DCM on the nervous system. Human volunteers were exposed to DCM via inhalation at average concentrations of 0, 317, 470, or 751 ppm for up to 4 h. Decreased visual and auditory functions were observed at 300 ppm or above, and decreased performance in most psychomotor tasks was demonstrated in groups subjected to 751 ppm compared with untreated controls. A second study (Stewart et al. 1972) reported impairment of the central nervous system (CNS) in two of three test subjects who inhaled vapors of DCM (1,000 ppm) for 1 to 2 h. The subjects demonstrated a reduced ability to respond to stimuli. A concentration of 500 ppm for a corresponding exposure duration was without effect. Central nervous system. Inhalation (animal). There are few studies on the effects of DCM on the CNS apart from experiments that quantify the anesthetic response. The percentage of sleeping time characterized by rapid eye movements was reduced in rats following exposure to 1,000 ppm for 24 h (Fodor and Winneke 1971). At 5,000 to 9,000 ppm, long sleeping periods occurred without the desynchronization phases that usually appear every few moments in normal sleep (Berger and Fodor 1968). Exposure to 5,000 ppm has been shown to reduce the spontaneous running activity of rats (Heppel et al. 1944). Central nervous system, inhalation (oral and dermal). No studies were found in the available literature on humans and animals. Hepatotozicity, inhalation (human). No data were found in the available literature on acute DCM-Induced hepatotoxicity in humans. Long-term studies involving occupational exposures did suggest qualitatively that DCM may cause liver toxicity, as evidenced by a reported case of hepatitis and altered triglyceride levels. The importance of these responses is reduced since no supporting quantitative data were provided. ------- Health Effaces Summary 17 Hepatotozicity, Inhalation (animal). Structural and biochemical changes in the liver have been reported following short- and long-term inhalation exposure to DCM. Cytoplasmic vacuolization and/or fatty changes were observed in the livers of mice, rats, dogs, and monkeys following continuous inhalation exposures in the range of 25 to 1.000 ppm for 100 days (Haun 1972). Rats and dogs showed mild effects at concentrations as low as 25 ppm; mice exhibited fat accumulation and glycogen depletion at continuous exposure to 100 ppm, and microscopic examination of the livers of monkeys exposed to 1,000 ppm showed mild fatty degeneration. Further, fatty changes were also observed in dogs continuously exposed to DCM (5,000 ppm) for 25 h (HacEwen et al. 1972) and in mice at 5,200 ppm for 6 h (Morris et al. 1979). Rats were exposed to DCM via inhalation for 2 years at concentrations of 50, 200, and 500 ppm (Nitschke et al. 1982, 1988), or at 500, 1,500, and 3,500 ppm (Burek et al. 1980. 1984). Hepatic vacuolization was noted in both sexes at 500 ppm. The incidence of multinuclear hepatocytes was also increased in females at exposures greater than 500 ppm. Higher exposure levels resulted in increased numbers of foci, or areas of altered hepatocytes (at 3,500 ppm in females), and necrosis (at 1,500 ppm in males and 3,500 ppm in females). A third study (NTP 1986) reported hemosiderosis, hepatomegaly, cytoplasmic vacuolization, and focal necrosis at all test doses in rats of both sexes exposed to 1,000, 2,000, and 4,000 ppm DCM for 2 years. In studies with mice (NTP 1986), hepatic cytologic degeneration was apparent at 4,000 ppm in both sexes and at 2,000 ppm in females. Hepatotozicity, oral (human). No hepatotoxicity studies were found in the available literature on the oral administration of DCM in humans. Hepatotozicity. oral (animal). No data were found in the available literature on the histomorphological effects of DCM in the liver following acute oral exposure. There is, however, some evidence for biochemical changes. The microsomal cytochrome P-450 content in rats was reduced 18 h after being dosed with 1 g/kg DCM (Moody et al. 1981). Increased triglyceride content, reduced triglyceride secretion, and reduced tubulin protein content were reported for mice given 2.7 g/kg DCM. Prolonged exposure to DCM produced mild toxic effects in the liver. The National Coffee Association (NCA 1982) evaluated the chronic toxicity of DCM ingested by F344 rats (50/sex/dose) in drinking water at target doses of 0, 5, 50, 125, or 250 mg/kg/day for 104 weeks. Additional high-dose (250 mg/kg/day) groups (25 animals/sex) were administered DCM for 78 weeks and then allowed to recover for a period of 26 weeks. Hepatic histological alterations, including increased incidence of foci (areas of cellular alterations), were detected at 78 weeks and at 104 weeks of treatment in the 50-, 125-, and 250-mgAg/day dose groups for both sexes. Fatty liver changes occurred in rats of both sexes given 125 and 250 mg/kg/day. A no-effect level of 5 mg/kg/day was determined. Male and female B6C3F1 mice exposed to 0, 60, 125, 185, or 250 mg/kg/day of DCM in their drinking water for 2 years exhibited liver effects only at the highest dose (NCA 1983). A slight increase in the incidence of foci of altered morphology (hepatocellular hyperplasia) occurred in treated males, but the effect was not statistically ------- 18 Section 2 significant; the number of animals with this effect was within the normal variation for this strain of mouse. An increased amount of fatty change occurred in mice given 250 mg/kg/day. A no-effect level of 185 mg/kg/day was determined. Hepatotoxicity. oral (dermal). No hepatotoxicity studies were found in the available literature on the dermal administration of DCM in humans and animals. Renal effects, inhalation (human). No association between DCM exposure and adverse effects on the kidney were reported in several epidemiologic studies (Friedlander et al. 1978, Ott et al. 1983, Hearne et al. 1987). Renal effects, inhalation (animal). Continuous exposure to DCM (25 and 100 ppm) for 100 days showed nonspecific renal tubular degenerative and regenerative changes in rats; however, there were no changes in the organ-body weight ratio. A NOAEL of 100 ppm for 100 days was determined for the dog (Haun et al. 1972). Renal effects, inhalation (oral and dermal). No studies were found in the available literature on renal toxicity following the oral and dermal administration of DCM in humans and animals. 2.2.1.3 Developmental toxicity No data were found in the available literature on the effects of DCM in humans following inhalation, oral, or dermal exposure. Animal data demonstrated that inhalation of DCM at dose levels of 1,250 ppm produced a minor skeletal variant (extra sternebrae, P < 0.05) in mice (Schwetz et al. 1975). Fetus weight was reduced, and behavioral changes occurred in rat pups following exposure of dams to 4,500 ppm DCM (Hardin and Manson 1980). The practical importance of these findings is limited. since each of the two studies used only one dose level and the observed effects occurred at maternally toxic doses. Evidence, therefore, does not currently exist to conclusively characterize the developmentally toxic potential of DCM in humans. 2.2.1.4 Genotoxicity Methylene chloride has been evaluated in a variety of short-term mammalian systems in vitro and in vivo to assess its potential to induce gene mutation and cause chromosomal aberrations and DNA/damage and repair (I11ing and Shlllaker 1985, CEFIC 1986d). Methylene chloride has been shown to cause mutations in in-vitro test systems employing bacteria or yeast. Results were generally negative in vivo. Overall, DCM appears to be a weak mutagen in lower species, but evidence of mutagenicity in mammalian cells is lacking. 2.2.1.5 Reproductive toxicity Methylene chloride did not adversely affect reproduction in rats exposed to 0. 100, 500, or 1,500 ppm via inhalation for two generations (Nitschke et al. 1985, unpublished). ------- Health Effaces Summary 19 2.2.1.6 Carcinogenicity Inhalation, human. Epidemiologies! studies have not demonstrated a significant increase in cancer deaths Ln humans occupationally exposed to DCM. The results of studies of deaths in male workers exposed to TWA concentrations (30 to 125 ppm) for up to 30 years showed no excess liver or lung cancer mortality (Frledlander et al. 1978). No excess deaths from lung and liver cancer were reported in a study evaluating the mortality experiences of an expanded cohort exposed at a rate of 26 ppm (8-h TWA) for 22 years (median latency of 30 years) (Hearne et al. 1987). However, an elevated incidence of pancreatic cancer deaths was reported in the Hearne et al. 1987 study. These deaths were not considered to be statistically significant. Ott et al. (1983a) evaluated cancer mortality among employees of a fiber production plant who were exposed to TWA concentrations of 140 to 475 ppm. They observed fewer than expected deaths from malignant neoplasms in comparison to expected deaths from the same cause in the general population. Although the available human evidence suggested that DCM is not carcinogenic at levels of exposure in an occupational setting of up to 475 ppm, these epidemiological studies were inadequate and cannot rule out the possibility of some risk associated with exposure to DCM. Based on animal studies, EPA considers DCM to be a probable human carcinogen (EPA 1987b). Inhalation, animal. Methylene chloride induced a dose-dependent, statistically significant increase in liver and lung adenomas and carcinomas in male and female mice exposed via inhalation for a lifetime at concentrations of 2,000 or 4,000 ppm (NTP 1986). Tumor incidences were as follows: at 2,000 ppm, 30 of 48 female mice and 27 of 50 male mice developed lung tumors compared with 3 of 50 female mice and 5 of 50 male mice in controls. Female mice (16 of 48) and male mice (24 of 49) also developed liver tumors. At 4,000 ppm, 41 of 48 female mice and 40 of 50 male mice developed lung tumors; 40 of 48 female mice and 33 of 49 male mice developed liver tumors. In controls, liver tumors developed in 3 of 50 female mice and in 22 of 50 male mice. In a rat bioassay (NTP 1986), DCM induced a statistically significant increase in benign mammary tumors, of a type not expected to progress into malignant tumors (McConnell et al. 1986), in female rats exposed at 2,000 or 4,000 ppm. Male rats developed mammary gland fibroadenomas at 4,000 ppm, but only at a marginally significant rate. The NTP interpreted its study as showing clear evidence of animal carcinogenicity. Oral, human. No carcinogenicity studies were found in the available literature on oral administration in humans. Oral, animal. A carcinogenic effect has been demonstrated in laboratory animals following long-term oral exposure to DCM. The National Coffee Association (NCA) evaluated the carcinogenic potential of DCM in rats and mice. In the rat study (NCA 1982), DCM (at doses of 0, 5, 50, 125, and 250 mgAg/day) was administered in drinking water to Fischer 344 rats of both sexes for 104 weeks. An increase in liver tumors, which was within historical control incidences, was noted in females treated at 50 and 250 mg/kg/day when compared to concurrent ------- 20 Section 2 controls. The authors did not consider these effects to be attributed to DCM treatment, since there was an unusually low incidence of similar tumors in the concurrent control groups and an absence of increased incidence of hepatic tumors in the group treated at 125 mg/kg/day. The EPA (1985a) evaluated the results of this study and reported an increased incidence of neoplastic nodules combined with hepatocellular carcinomas in female rats (P < 0.05) when compared with matched controls. Tumor incidences were as follows: 0/134, 1/85, 4/83, 1/85, and 6/85 in combined control, 5-, 50-, 125-, and 250-mg/kg/day groups, respectively. The EPA (1985a) concluded that based on the combined incidence of adenomas and carcinomas, DCM showed borderline evidence of careinogenieity in rats when administered orally. In the mouse study (NCA 1983), DCM was administered in drinking water at doses of 0, 60-, 125-, 185-, and 250-mgAg/day for 104 weeks. A slight increase (not statistically significant) in proliferative hepatocellular lesions was reported in treated male groups when compared with male controls (but not in females). The authors concluded that DCM was not carcinogenic under conditions of this study, since the tumor incidences were not dose related and were within historical control levels. An EPA (1985b) assessment of the mouse study reported a marginally significant (P < 0.05) increase in the combined incidence of hepatocellular adenoma and carcinoma in male mice (24/125, 51/200, 30/100, 31/99, and 35/125 in combined control, 60-, 125-, 185-, and 250-mgAg/day groups, respectively). The EPA (1985b) considers the evidence for the carcinogenicity of DCM in mice via oral exposure to be borderline. 2.2.1.7 Mechanisms of action The primary effect produced following DCM exposure was impairment of the CNS. Methylene chloride also affected the cardiovascular system, the liver and kidneys, was mutagenic in bacteria and yeast, and produced tumors in laboratory animals. Adverse effects produced following DCM exposure may be due to the parent compound, its reactive intermediates, or a combination of these. The CNS toxicity of DCM may be attributable to direct central depressant action, since DCM is a known anesthetic agent. This effect would be expected to be important if high-level exposures were encountered. Methylene chloride undergoes metabolism to reactive intermediates which become irreversibly bound to tissue protein and lipids. The relationship between this bioactivation and the fatty liver produced by DCM is not clear, but metabolism appears to be involved in the mutagenic effect of DCM seen in bacteria and yeast. The CNS toxic icy of DCM may also be associated with the carbon monoxide produced as a metabolite of DCM. Carbon monoxide may be potentially dangerous in individuals with cardiovascular disease, and may contribute to deficits in human performance detected by behavioral testing following DCM exposure. Based on studies in mice. DCM's carcinogenicity is most likely produced as a result of metabolism via the glutathione conjugation [glutathione-S-transferase (GST)] pathway. Parent DCM and the oxidation ------- Health Effects Summary 21 [mixed-function oxidases (MFO)] pathway are considered to be possible sources of tumorigenic potential rather than probable sources. Neither the parent nor the oxidation pathway correlates with tumor incidence, either among species or across doses. Current evidence is not sufficient to identify with reasonable certainty the carcinogenic mechanism of action of DCM. Genotoxicity and cytotoxicity have been investigated; however, neither has been clearly shown to be the cause of carcinogenicity seen in animal studies. Additional data on the mechanism of action may be provided by the National Institute of Environmental Health Science (NIEHS), which plans to investigate further the role of cell replication in DCM tumorigenesis and the pattern of oncogene activation in spontaneous and dose-related tumors. Also, the European Center of Chemical Manufacturers' Federation (CEFIC) is conducting studies to evaluate the effects of DCM in Clara cells in relation to tumor development. 2.2.2 Biological Monitoring as a Measure of Exposure and Effects Methylene chloride exposure may be monitored by its determination in the blood, breath, or urine of exposed persons (NIOSH 1976). Methylene chloride and formic acid were found in measurable amounts in the urine of persons exposed to the chemical. The amount of DCM found in a 24-h period following a 24-h exposure at 100 and 200 ppm was proportional to the exposure concentrations (DiVincenzo et al. 1972). Formic acid was found in the urine of most persons exposed to DCM in a particular occupational study, but no correlation between the intensity of exposure and the concentration of formic acid could be determined (Kuzelova and Vlasak 1966). Methylene chloride has also been measured in the blood and breath of exposed persons. The concentrations of DCM in the blood and breath have been representative of the exposure (Astrand et al. 1975, Riley et al. 1966, DiVincenzo et al. 1972). Carboxyhemoglobin (CO-Hb) in persons exposed to DCM at rest is dependent on the exposure concentration, duration, and the extent of concurrent carbon monoxide exposure. These relationships, however, are disrupted by physical activity. With increased activity, maximum CO-Hb values may not be attained until 3 to 4 h after the end of the exposure. Although it is possible to monitor CO-Hb levels in workers exposed to DCM, the time that the measurements should be made in relation to the end of exposure has not been well established. Consequently, a series of measurements may prove more informative. Since the relationship between alveolar carbon monoxide and CO-Hb has not been well established for DCM workers, breath analysis for carbon monoxide cannot be considered as providing definitive quantitative information regarding DCM exposure. 2.2.3 Environmental Levels as Indicators of Exposure and Effects 2.2.3.1 Levels found in the environment Methylene chloride has been detected in ambient air, in surface and drinking water, and at very low levels in decaffeinated coffee and spices. However, these sources of exposure are considered to be minor. ------- 22 Section 2 Mechylene chloride occurs in ambient air with levels in the parts-per- trillion range and has been detected only at low levels (7 mg/L was the maximum level) in surface and drinking water (EPA 1980). The U.S. Food and Drug Administration reports that DCM levels in decaffeinated coffee and spices are extremely low. Methylene chloride is also formed from natural sources; however, natural sources are not considered as significant contributors to environmental concentrations (EPA 1980). 2.2.3.2 Human exposure potential Exposures to DCM occur primarily in workplaces that use DCM and also through the use of DCM consumer products. Exposure to DCM in outdoor air, water, and food is low. 2.3 ADEQUACY OF DATABASE 2.3.1 Introduction Section 110 (3) of SARA directs the Administrator of ATSDR to prepare a toxicological profile for each of the 100 most significant hazardous substances found at facilities on the CERCLA National Priorities List. Each profile must include the following content: "(A) An examination, summary, and interpretation of available toxicological information and epidemiologic evaluations on a hazardous substance in order to ascertain the levels of significant human exposure for the substance and the associated acute, subacute, and chronic health effects. (B) A determination of whether adequate information on the health effects of each substance is available or in the process of development to determine levels of exposure which present a significant risk to human health of acute, subacute, and chronic health effects. (C) Where appropriate, an identification of toxicological testing needed to identify the types or levels of exposure that may present significant risk of adverse health effects in humans." Thia section identifies gaps in current knowledge relevant to developing levels of significant exposure for DCM. Such gaps are identified for certain health effect end points (lethality, systemic/target organ toxicity, developmental toxicity, reproductive toxiclty, and carcinogenicity) reviewed In Sect. 2.2 of this profile in developing levels of significant exposure for DCM. and for other areas such as human biological monitoring and mechanisms of toxicity. The present section briefly summarizes the availability of existing human and animal data, identifies data gaps, and summarizes research in progress that may fill such gaps. Specific research programs for obtaining the data needed to develop levels of significant exposure for DCM will be developed by ATSDR, NTP, and EPA in the future. ------- Health Effects Summary 23 2.3.2 Health Effect End Points 2.3.2.1 Introduction and graphic summary Because DCH Is found in consumer products such as insecticides, metal cleaners, paints, and paint varnish removers, and occurs in water, numerous evaluations have been conducted to assess its potential as a human health hazard. The availability of data for health effects in humans and animals is depicted on bar graphs in Figs. 2.5 and 2.6, respectively. The bars of full height indicate that there are data to meet at least one of the following criteria: 1. For noncancer health end points, one or more studies are available that meet current scientific standards and are sufficient to define a range of toxicity from no-effect levels (NOAELs) to levels that cause effects (LOAELs or FELs). 2. For human carcinogencity, a substance is classified as either a "known human carcinogen" or "probable human carcinogen" by both EPA and International Agency for Research on Cancer (IARC) (qualitative), and the data are sufficient to derive a cancer potency factor (quantitative). 3. For animal carcinogenicity, a substance causes a statistically significant number of tumors in at least one species, and the data are sufficient to derive a cancer potency factor. 4. There are studies which show that the chemical does not cause this health effect via this exposure route. Bars of half height indicate that "some" information for the end point exists but does not meet any of these criteria. The absence of a column indicates that no information exists for that end point and route. 2.3.2.2 Description of highlights of graphs Most of the studies in the available literature are for inhalation exposure; an exception is a 2-year drinking water study with rodents. Systemic effects have been adequately evaluated in animals; similarly, such effects have been observed in humans, but the reported findings were not supported by quantitative and histomorphological data. 2.3.2.3 Suaaary of relevant ongoing research Several studies are in progress that would contribute new and useful Information on DCM toxicity. These studies include the following • Role of cell replication in DCM tumorigenesis and the pattern of oncogene activation in spontaneous and dose-related tumors (by NIEHS). • Liver cell turnover as a mechanism of carcinogenesis in mouse liver (by CEFIC). ------- HUMAN DATA ro -P- ZIZ J SUFFICIENT 'INFORMATION* J SOME INFORMATION NO INFORMATION ORAL INHALATION DERMAL LETHALITY ACUTE INTERMEDIATE CHRONIC DEVELOPMENTAL REPRODUCTIVE CARCINOOENICITY ZL _ y TOXICITY TOXICITY SYSTEMIC TOXICITY 'Sufficient information exists to meet at least one of the criteria for cancer or noncancer end points. Fig. 2.5. Avuiliibilily of information on health effects of melhylene chloride (human data). ------- ANIMAL DATA v SUFFICIENT "INFORMATION* J . SOME INFORMATION NO INFORMATION ORAL INHALATION DERMAL LETHALITY ACUTE INTERMEDIATE CHRONIC DEVELOPMENTAL REPRODUCTIVE CARCINOOENICITY / / TOXICITY TOXICITY 3 m n PI in fVSTEMIC TOXICITY 'Sufficient information exists to meet at least one of the criteria for cancer or noncancer end points. Kig. 2.6. Availability of information on health effects of methylene chloride (animal data). ro t-n ------- 26 Section 2 • Glutathione pathway rates from the 36C1 isotope (by Dow Chemical Company and CEFIC). • Measurements of new partition coefficients (by CEFIC). • In vivo pathway measurements with deuterated DCM (by CEFIC). These studies will help elucidate the mechanism of action of DCM and will improve knowledge of species differences in pharmacokinetics. 2.3.3 Other Information Needed for Human Health Assessment 2.3.3.1 Pharmacokinetics and mechanisms of action The pharmacokinetics of DCM have been extensively studied. Based on these studies, two metabolic pathways are known: one involving glutathione-S-transferase (GST) and the other involving oxidation mixed-function oxidase (MFO). The GST pathway is believed to produce carbon dioxide (C02), whereas the MFO pathway produces CO and C02. The current evidence is not sufficient to identify with reasonable certainty the mechanism of action of DCM. Based on studies in rodents (mice), DCM's carcinogenicity is believed to be the result of metabolism via the GST pathway. Both pathways may be active in mice at low doses, but at higher doses the MFO pathway becomes saturated, and the metabolic load is increasingly shifted to the alternative GST pathway. Parent DCM and the MFO pathway are considered to be possible sources of tumorigenic potential rather than probable sources. Neither one correlates with tumor incidence, either among species or across doses. Further, the parent compound is thought to be chemically unreactive. Much uncertainty exists about the mechanism of action of DCM. -The genotoxicity and cytotoxicity of DCM have been investigated as possible mechanisms of carcinogenic action, but neither has been shown definitively to be the cause for the tumorigenicity observed in animal studies. Additional data on mechanisms are needed. The NIEHS is investigating the role of cell replication in DCM tumorigenesis and the pattern of oncogene activation in spontaneous and dose-related tumors. The CEFIC is conducting studies to evaluate the effects of DCM in the Clara cell in relation to lung tumor development. Methylene chloride produces liver and kidney effects in laboratory animals; however, the mechanism by which these effects are produced is not known. The CNS effects produced following exposure to DCM are probably due to DCM alone or in combination with CO-Hb. 2.3.3.2 Monitoring of human biological samples Several methods are used to test human exposure to DCM. The analysis of DCM in breath is a good indicator of exposure since most of the DCM taken Into the body is eliminated unchanged In expired air from the lungs. Analysis of the blood and urine can also be used to determine exposure to DCM in humans. 2.3.3.3 Environmental considerations Current methodologies to assess the levels of DCM in the environment are adequate. However, there is an absence of adequate data on exposure from the ambient environment. ------- 27 3. PHYSICAL AND CHEMICAL INFORMATION 3.1 CHEMICAL IDENTITY The chemical formula, structure, synonyms, trade names, and Identification numbers for DCM are listed in Table 3.1. 3.2 PHYSICAL AND CHEMICAL PROPERTIES Important physical and chemical properties of DCM are listed in Table 3.2. ------- 28 Section 3 Table 3.1. Chemical identity of methylene chloride Chemical name Synonyms Trade names Chemical formula Wiswesser line notation Chemical structure Identification numbers: CAS Registry No. NIOSH RTECS No. EPA hazardous waste No. OHM-TADS No. DOT/UN/NA/IMCO Shipping No. STCC No. Hazardous Substances Data Bank No. National Cancer Institute No. Methylene chloride Dichloromethane DCM Methane dichlonde Methane, dichloro (8CI and 9CI) Methylene bichloride Methylene dichlonde Aerothene MM Freon 30 Narkotil R30 Solaesthin Solmethme CH2C12 GIG Cl H-C-H l Cl 75-09-2 PA8050000 U080, F002 7217234 UN 1593 66 C50102 Source: HSDB 1987. ------- Chemical and Physical Information 29 Table 3.2. Physical and chemical properties of methylene chloride Property Molecular weight Color Physical state Odor Odor threshold Melting point Boiling point Autoignition temperature Solubility Water (20 to 30° C) Organic solvents Value 84.93 Colorless Liquid Characteristic, sweetish, not unpleasant 1 50-600 ppm -95.1°C 40° C (at 760 mm Hg) 1,033°F 20,000 mg/L Miscible with a wide References Weast 1985 Verschueren 1977 Verschueren 1977 Verschueren 1977 Ruth 1986 Weast 1985 Weast 1985 NFPA 1984 EPA 1986a EPA I985c Density at 20° C Vapor density, air = 1 Partition coefficients Octanol-water (Kg,,) log Organic carbon (£«) Vapor pressure (20 and 30° C) Henry's law constant Refractive index (20°C) Flash point (closed cup) Flammable limits variety of organic solvents 1.3266 g/mL 2.93 Weast 1985 Verschueren 1977 Conv factors 1.30 EPA 1986a 8.8 g/mL EPA 1986a 349 and 500 mm Hg EPA 1986a 2.03 X 1CT3 atm-mVrnol EPA 1986a 1.4242 Weast 1985 Practically nonflammable NFPA 1984 14-22% NFPA 1984 1 ppm = 3.53 mg/m3 Verschueren 1977 in air ------- 31 A. TOXICOLOGICAL DATA 4.1 OVERVIEW Mechylene chloride is a volatile liquid with high lipid solubility and modest solubility in water. It is absorbed primarily by inhalation and ingestion. Dermal exposure has resulted in DCM absorption in laboratory animals and humans, but this occurs at a slower rate than when administration is by other routes. Consistent with the properties of high lipid solubility and modest water solubility, adsorption of DCM following inhalation and ingestion is rapid. Once absorbed, DCM is quickly distributed to a wide range of tissues and body fluids. Methylene chloride is metabolized via two pathways, the MFC and the GST. The GST pathway produces C02, whereas the HFO pathway produces CO and C02. Methylene chloride is almost exclusively metabolized by the MFO pathway at low exposures. Acute exposure to DCM has been associated with impairment in function of the central nervous system and liver and kidney effects. In human experimental studies, DCM (330 ppm) decreased visual and auditory functions and impaired psychomotor tasks (800 ppm) following inhalation exposure for 5 h. Reduced sleeping time was reported in rats exposed to DCM (1,000 ppm) for 24 h. Subchronic exposure to DCM resulted in mild liver and kidney effects in animals. Vacuolization and fatty changes in liver cells were reported in mice, rats, and dogs that inhaled DCM (100 ppm) for 100 days. Degenerative and regenerative changes in kidney tubules were also reported in rats exposed via inhalation to DCM (25 or 100 ppm) for 100 days. It should be noted that liver and kidney effects were elicited after continuous exposure without recovery as opposed to the more traditional exposure protocol. Chronic exposure to DCM has been associated with mild liver toxicity, as evidenced by cytoplasmic vacuolization, increased fat content, and multinucleated hepatocytes following inhalation exposure at >SOO ppm for 2 years. Histomorphological alterations and fatty changes were also noted in mice and rats following oral exposure for 2 years. Liver tumors were produced in rats and mice exposed to DCM in drinking water for 2 years; however, the tumor incidences were not considered by the study authors to be compound related since the incidences were within historical control levels. An EPA assessment of these responses reported borderline statistical increases in hepatic neoplastic nodules and carcinomas (combined) in rats and mice following ingestion of DCM in drinking water for 2 years. Liver and lung neoplasms in mice and benign neoplasms in the mammary gland in rats were reported following inhalation exposure to DCM for 2 years. In one study, sarcomas were observed in the salivary gland region in male rats following ------- 32 Section 4 inhalation of DCM for 2 years; however, these tumors have not been repeated in other bioassays on rats. Epidemiological studies found no association between DCM exposure and liver and lung tumors in humans. Based on the weight of evidence from animal studies, DCM was classified by EPA as a probable human carcinogen; however, metabolic data pointing to species differences in the utilization of the DCM metabolic pathways indicate that risks to humans are lower than those determined for laboratory animals. 4.2 TOXICOKINETICS 4.2.1 Overview Methylene chloride is a small lipophilic molecule that is rapidly absorbed from the alveoli of the lung into the systemic circulation. It is also readily absorbed from the gastrointestinal (GI) tract. Following GI tract absorption, DCM may be subject to first-pass hepatic metabolism and elimination before reaching the systemic circulation. Dermal exposure to DCM also results in absorption but at a slower rate than other exposure routes. Absorption and distribution of DCM can be affected by a number of factors, including dose level, dose vehicle, physical activity, duration of exposure, and amount of body fat. Methylene chloride is metabolized in vivo via two pathways: (1) an oxidative (MFC) pathway mediated by the P-4SO system that yields CO and C02 and (2) a glutathione-dependent (GST) pathway that only yields C02- The MFO pathway is saturable at air concentrations of a few hundred parts per million. However, the GST pathway shows no indication of saturation at inhaled concentrations of up to 10,000 ppm. In vitro studies have also demonstrated two metabolic pathways. An oxidative pathway mediated by the P-450 system yields CO, and a second pathway mediated by a glutathione-dependent reaction yields formaldehyde and formic acid, which are further metabolized to C02. Elimination of DCM from the body is primarily via expired air from the lungs as unchanged parent compound or as CO and C02, the major metabolites. The dose is a major determinant of the elimination product. At low doses of DCM, a large percentage of the administered dose is metabolized to CO and C02. However, the percentage of administered dose that is metabolized is reduced as the dose of DCM is increased. In this case, more of the unchanged parent compound is exhaled in expired air. A small fraction of absorbed DCM has been detected in urine and feces. The results of recent pharmacokinetic studies suggest that the parent compound and/or reactive metabolites produced by the GST pathway are the source of DCM-induced carcinogenicity. Based on these studies, it has been postulated that the activity of the GST pathway in humans Ls less than that of mice and might only become significant when the P-450 pathway has been saturated. Because of its low chemical reactivity, DCM itself is unlikely to be directly Involved in carcinogenesis. Consequently, metabolism of DCM by GST appears to be important in carcinogenesis. Recent studies indicate that the GST pathway is less active in rats, hamsters, and humans than in mice. ------- ToxicologLcal Daca 33 4.2.2 Absorpt ion 4.2.2.1 Inhalation Human. The principal route of human exposure to DCM is inhalation. Absorption is rapid, followed by a plateau of blood concentration in several hours. At low levels of exposure, blood concentrations of DCM increase linearly with exposure level. However, at high concentration levels, saturation occurs. Riley et al. (1966) reported that in one test subject exposed to 100 ppm DCM for 2 h, the amount of DCM absorbed was initially 70%, but gradually decreased to 31% of the DCM content of inspired air. Similarly, 70 to 75% of inhaled vapor of DCM was absorbed in human subjects exposed by inhalation to 50, 100, 150, or 200 ppm (174. 347, 521, or 694 mg/m3) DCM for 7.5 h on a single occasion (DiVincenzo and Kaplan 1981). The resulting pulmonary uptake was found to be 5.54, 10.70, 15.38, and 21.07 mmol (471, 909, 1,306, and 1,790 mg), respectively. The percent absorption was relatively constant over the range of exposures evaluated. McKenna et al. (1980) exposed volunteers to 100 or 350 ppm (347 or 1,215 mg/m3) of DCM for 6 h. Their data showed that the blood level of DCM for both concentrations reached a steady state in -2 h. The amount of DCM absorbed increased with duration of exposure and physical activity (resulting in increased ventilation and cardiac output). Physical activity for 0.5 h during exposure to 250 or 500 ppm DCM doubled absorption but decreased retention from 55 to 40% because of a threefold (6.9 to 22 L/min) increase in ventilation rate (Astrand et al. 1975). Methylene chloride absorption was also related directly to the degree of obesity in human subjects (Engstrom and Bjurstrom 1977). Obese subjects absorbed 30% more DCM than lean subjects when exposed to 75 ppm for 1 h. Animal. The magnitude of DCM uptake depends,on several factors, including inspired air concentration, pulmonary ventilation, duration of exposure, the rates of diffusion into blood and tissues, and solubility in blood and the various tissues. The concentration of DCM in alveolar air, in equilibrium with pulmonary venous blood content, asymptotically approaches the concentration in the inspiratory air until a steady-state condition is reached (EPA 1985a). After tissue and total body steady state is reached during exposure, uptake is balanced by elimination through the lungs and other routes, including metabolism. DiVincenzo et al. (1972) investigated the absorption of DCM (99% purity) in dogs (fasted male beagle, number of animals per dose not specified) exposed by inhalation to 100, 200, 500, or 1,000 ppm (347, 694, 1,735, or 3,470 mg/m3) DCM for 2 or 4 h. Expired air and blood concentrations of DCM were reportedly proportional to the magnitude of exposure. Absorption of DCM was reportedly a function of its solubility in blood and tissues, the cardiac output, and respiratory rate. No quantitative data were presented to support the authors' conclusion. MacEwen et al. (1972) determined blood DCM concentrations to be directly proportional to exposure concentrations when dogs exposed for 16 days to 1.000 ppm (3,474 mg/m3) and 5,000 ppm (17.370 mg/m3) exhibited blood DCM levels of 36 and 182 mg/L, respectively. Total equilibrium can be assumed to have occurred in these animals (EPA ------- 34 Section 4 1985a). Similar values can be calculated from the data of Latham and Potvin (1976). They found a proportional relationship in rats between DCM blood and inspired air concentrations over a range of 1,000 to 8,000 ppm (3,474 to 27,792 mg/m3) during a 6-h exposure. In contrast to these findings of a direct proportional relationship between inspired air concentration of DCM and blood level in man and other animals, McKenna et al. (1982) reported a greater than proportionate increase of blood concentration with inhalation exposure concentration in rats. Male Sprague-Dawley rats were exposed for 6 h to 50, 500, and 1,500 ppm DCM (173, 1,737 and 5,211 mg/m3). The data from McKenna et al. (1982) indicate that the whole blood and plasma levels of DCM increase disproportionately with an increase in inspired air concen- tration. Furthermore, calculation of the blood/air ratio for these data provide increasing values of 5.75, 5.97, and 7.59 for 50, 500, and 1,500 ppm, respectively. McKenna et al. (1982) suggest that the resultant increase in blood DCM concentration is greater than that predicted by increments in the inspired air because of a rate-limited metabolism of DCM in the rat. Thus, at low inspired air concentrations (below saturation of metabolism), the blood/air ratio is less (because of rapid metabolism) than that at high inspired air concentrations (above saturation of metabolism). At the highest dose level, the blood/air coefficient is in agreement with values reported by other investigators. 4.2.2.2 Oral Human. No studies were found in the available literature on the oral absorption of DCM in humans. Animal. Limited direct absorption studies in rats and mice demonstrated that DCM was readily absorbed from the gastrointestinal tract. Methylene chloride levels detected in gut segments up to 40 min posttreatment were similar between doses in rats administered single oral doses of nonradioactive DCM (in water) at 50 or 200 mgAg (Angelo et al. 1986a). Sixty percent of the administered dose (200 mgAg) was recovered from the upper gastrointestinal tract <10 min posttreatment (20% recovery after 40 min). The amount of DCM in the lower gastrointestinal tract accounted for <2% of the administered dose up to the 40-min test interval. In mice administered oral doses of nonradioactive DCM at 10 or 50 msAg (in water), -25% of the administered dose was detected in the upper gastrointestinal tract within <20 min (Angelo et al. 1986b). Similarly, after treatments with DCM at 10, 50, or 1,000 mg (in corn oil), -55% of the administered dose was detected in the upper gut segment and remained there 2 h (Angelo et al. 1986b). Reported findings that DCM is metabolized to CO and C02 and that unchanged DCM is eliminated in the breath are in support of findings from limited direct absorption studies demonstrating the absorption of DCM when ingested. A single oral dose of 1 or 50 mgAg ^C-DCM administered to rats was eliminated in the breath as unchanged DCM (12 3 or 72.1%, respectively) and as metabolites (primarily CO and O>2 at 88 and 28%, respectively) within 48 h (McKenna and Zempel 1981). Mice ------- lexicological Data 35 excreted 40% of the administered dose (100 mg/kg) in expired air as unchanged DCM and 45% as metabolites CO and C02 within 96 h (Yesair et al. 1977). 4.2.2.3 Dermal Human. No studies were found in the available literature on the dermal absorption of DCM in humans. Animal. Data show that DCM can be absorbed through the skin of laboratory animals. Maksimov et al. 1977 (cited in NIOSH 1976) immersed two-thirds of the tail of 128 white rats and measured the DCM concentration in various tissues (lung, liver, brain, kidney., heart, and fat) by gas chromatography after 1-, 2-, 3-, and 4-h exposures. Small increases were seen in most tissues after 1 or 2 h of exposure, and DCM concentrations in fatty tissues increased markedly after 3 h of exposure. After 4 h of exposure, DCM concentrations remained elevated in fatty tissues and were increased in all other tissues studied. 4.2.3 Distribution 4.2.3.1 Inhalation Human. There is some evidence for accumulation of DCM in body fat. However, it appears to reach steady state and wash out rapidly. Engstrom and Bjurstrom (1977) exposed 12 male subjects (6 slim and 6 obese) to 750 ppm of DCM (2,600 mg/m3) for 1 h. The total uptake of DCM was 1,116 ± 34 mg by the slim group and 1,445 ± 110 mg by the obese group. On a milligram-per-kilogram basis, the uptake was IS.6 mg/kg for the slim group and 15.0 mg/kg for the obese group. Needle biopsies showed that the adipose tissue contained -8 to 35% of the average total uptake for both groups. The amount of DCM absorbed correlated highly with the degree of obesity and body weight. In the six slim subjects, the concentration in the adipose tissue during the 4-h period after exposure was approximately twice that of the six obese subjects. However, despite lower concentrations, the obese subjects had a greater calculated amount of DCM in the total fat depots of the body. McKenna et al. (1980) exposed volunteers to 100 or 350 ppm of DCM for 6 h and measured blood and exhaled air levels of DCM, CO-Hb, and exhaled CO. At the end of the 6-h exposure, the CO-Hb concentration of the group exposed to 350 ppm of DCM was 1.4-fold higher than that of the group exposed to the lower dose. Likewise, the concentration of exhaled CO in the high-dose group was 2.1-fold higher than that of the group exposed* to 100 ppm. McKenna et al. (1980) concluded that their finding of a nonllnearity between administered dose and the CO-Hb and CO levels is an indication that metabolic saturation was approached. Animal. In rats exposed to 14C-DCM at 500 ppm (1,735 mg/m3) for 1 h, radioactivity was detected in the liver, brain, and fatty tissue. Concentrations fell by 25, 75, and 90%, respectively, at 2 h following a 1-h compound exposure (Carlson and Hultengren 1975). Following the exposure of rats to air containing 200 ppm DCM for 6 h/day for 5 days, tissue concentrations were measured in the brain, blood, liver, and perirenal fat on the fifth day of exposure after ------- 36 Section 4 0 (18-h exposure on day 4), 2, 3. 4, and 6 h of exposure (Savolainen et al. 1977). Although no absorption factors were discussed, the persistence in perirenal fat before exposure on the fifth day indicated considerable retention in fat relative to other tissues. 4.2.3.2 Oral Human. No distribution studies were found in the available literature on the oral administration of DCM in humans. Animal. In tissue distribution studies in which male Sprague- Dawley rats were administered single oral (gavage) doses of DCM (14C-DCM at 1 or 50 mg/kg), the highest concentration of radioactivity was detected in the liver, kidney, and lung and the lowest in fat. Tissue 14C activity represented primarily metabolites (McKenna and Zempel 1981). Methylene chloride administered orally to rats in single doses of 50 or 200 mg/kg over a period of 14 days was detected in the blood. liver, and carcass (Angelo et al. 1986a). The average levels of DCM decreased rapidly in the blood and liver between 10 and 240 min, but the maximum carcass concentration occurred at the intermediate 30-min time point. Methylene chloride concentrations in blood were generally less than proportional to dose, whereas liver and carcass concentrations were approximately proportional to dose. In studies with mice, DCM was administered by gavage in a water vehicle at 50 mg/kg for 14 days (Angelo et al. 1984b). Four hours following dosing, all of the DCM concentrations in the blood and in most tissue samples were below the limit of detection. Between days 1 and 14, the DCM concentration in the 10-min blood samples tended to decrease, whereas the levels at the same time point increased for the liver and remaining carcass. Following a 500- and 1,000-mg/kg treatment in corn oil, the rate of decline of DCM in the blood was slower than was observed for the treatment in water Blood concentrations between treatments, which were approximately proportional to dose when compared at the same time points as DCM concentrations in the liver and carcass, also decreased more slowly following administration in corn oil rather than in water. However, there was no consistent pattern in the elimination profiles for a given dose treatment when tissue concentrations at similar time points were compared between days during the course of the 14-day dosing schedule, especially for the liver. 4.2.3.3 Dermal Human. No distribution studies were found in the available literature on the dermal administration of DCM in humans. Animal. No distribution studies were found in the available literature on the dermal administration of DCM in animals. 4.2.4 Metabolism 4.2.4.1 Inhalation Human. Several groups of investigators have studied the formation of CO and CO-Hb in human subjects exposed to DCM. Carboxyhemoglobin ------- Toxicological Daca 37 concentrations (calculated from measurements of CO in exhaled air) were measured in four male plastic film workers (20 to 33 years old, two nonsmokers, two smokers agreed to abstain) exposed to 180 to 200 ppm (625 to 694 mg/m3) of DCM for 8 h (Ratney et al 1974). The CO-Hb concentration, expressed as percent saturation of hemoglobin with CO, was about 4.5% saturation at the beginning of the workday and rose to about 9% saturation after 8 h of exposure. The 24-h time-weighted CO-Hb concentration was 7.3% saturation. Astrand et al. (1975) exposed 14 human subjects (males 19 to 29 years old) to DCM by inhalation and measured arterial CO-Hb concentrations during exposure and for up to 2 h after the end of the exposure. The concentration of CO-Hb increased both during and after exposure. The authors reported that a concentration of about 5.5% saturation was reached with an exposure to 500 ppm (1,750 mg/m3). The observations by Stewart et al. (1972) that human subjects (11 males, aged 23 to 43 years, nonsmokers) exposed by inhalation to 500 to 1,000 ppm (1,735 to 3,470 mg/m3) DCM (99.5% pure) for 1 or 2 h experienced elevated CO-Hb concentrations suggested that DCM was metabolized to CO. The CO-Hb concentrations rose to an average of 10 1% saturation 1 h after the exposure of three subjects to 986 ppm (3,421 rag/m3) DCM for 2 h. The mean CO-Hb concentration (3.9% saturation at 17 h postexposure) remained elevated above the preexposure baseline value (1 to 1.5% saturation). The exposure of eight subjects to 515 ppm (1,787 mg/m3) DCM for 1 h increased the CO-Hb level, which also remained elevated above baseline for more than 21 h. Peterson (1978) exposed human subjects (11 males aged 20 to 39 years and 9 females aged 20 to 41 years) by inhalation to 50, 100, 250, or 500 ppm (174, 347, 868, or 1,735 mg/m3, chemical purity not specified) DCM for 1, 3, or 7.5 h for up to 5 successive days per week for 5 weeks. It was found that (1) CO-Hb concentrations could be predicted from DCM exposure parameters (but the breath concentrations of DCM correlated better with exposure parameters that did CO-Hb concentrations), (2) no differences in DCM metabolism between male and female subjects were detectable, and (3) no acceleration of DCM metabolism to CO occurred during exposure for 5 weeks to concentrations ranging from 100 to 500 ppm (347 or 1,735 mg/m3) DCM. Elevated urinary formic acid contents have been reported in film workers exposed by inhalation to DCM, indicating that DCM is metabolized to formic acid by humans (Kuzelova and Vlasak 1966, cited in NIOSH 1976). Formic acid was present in urine samples from most (number not specified) of the 33 film workers exposed for an average of 2 years to contaminated air. The authors reported that concentrations in the workplace air systematically exceeded 0.5 mg/L (500 mg/m3) DCM and occasionally exceeded 1.75 mg/L (17,500 mg/m-*). Animals. Fodor et al. (1973) observed that albino rats (number of animals and sex not stated) exposed by inhalation to 0, 50, 100, 500, or 1,000 ppm (0, 174, 347, 1,735, or 3.470 mg/m3) DCM (purity not specified) for 3 h showed CO-Hb formation (0.4, 3.2, 6.2, 10.5, or 12 5% saturation, respectively), confirming the observation that DCM is metabolized to CO. Similarly, Carlson and Hultengren (1975) reported that Sprague-Dawley rats (ten male) exposed by inhalation to ^C ------- 38 Section 4 (1,935 mg/m3 for 1 h) produced l^C-CO, and the amount of radioactivity Ln CO extracted from the blood was highly correlated to the amount of CO-Hb formed in the blood. Male New Zealand rabbits (number not specified) exposed via inhalation to 5,000 to 10,000 ppm (17,350 to 35,700 mg/m3) DCM for 20 min showed elevated CO-Hb concentrations (Roth et al. 1975). The authors did not present specific values for the percent CO-Hb produced at each exposure; however, they did show results to demonstrate that increases in CO-Hb concentrations were both dose-dependent and time-dependent. CO-Hb concentrations reached a maximum in 2 to 3 h and returned to control values by 8 h. A linear dose-response relationship between inhalation of increasing concentrations of DCM and increasing CO-Hb was also observed when four rabbits were exposed to 1,270 to 11,520 ppm DCM (4,407 to 39,974 mg/m3, purity not specified) for a 4-week period (days per week and hours per day not specified). However, the authors reported that rabbits exposed to similar DCM concentrations had changes in CO-Hb levels that varied by up to a factor of 2. McKenna et al. (1982) reported that DCM is metabolized to CO and C02. In rats, inhalation of 50 ppm for 6 h resulted in 26 and 27% of the body burden being recovered as expired CO and C02, respectively, during the first 48 h after the end of the 6-h inhalation period. At 1,500 ppm, these numbers were 14 and 10%, respectively. A lesser percentage of the initial dose is metabolized to these end points at the high dose. Gargas et al. (1986) studied the in vivo metabolism of inhaled DCM in male Fischer 344 rats. They demonstrated that DCM displays complex uptake behavior, with both first-order and saturable components of metabolism. Gargas et al. (1986) compared plasma concentrations of CO-Hb in rats pretreated with pyrazole (which inhibits P-450 oxidation) or 2,3-epoxypropanol (2,3-EP) (which depletes glutathione) to assess the relative contribution of each pathway to the total metabolism of DCM. Pretreatment with pyrazole essentially abolished CO production by the high-affinity saturable P-450 pathway. Concentrations of CO-Hb following 2,3-EP pretreatment were increased 20 to 30% compared with untreated controls. Gargas et al. (1986) concluded that the effect of pyrazole pretreatment provides support for the involvement of cytochrome P-450 in the oxidation of DCM to CO. The in vivo pharmacokinetics of DCM and its major metabolites, CO and C02, were determined in F344 rats and B6C3F1 mice both during and immediately after a 6-h inhalational exposure to 500, 1,000, 2,000, and 4,000 ppm DCM (CEFIC 1986b). An assessment has been made of the relative utilization by the two species of the two known pathways of DCM metabolism. Maximum blood levels of DCM were reached within 3 h of the start of exposure and maintained at a constant level until the end of exposure when they declined rapidly in both species. The cytochrome P-450 pathway leading to CO and CO-Hb was shown to be saturated at the 500-ppm exposure level. After saturation of this pathway had occurred, the blood levels of DCM in the rat increased almost linearly with dose, indicating little further metabolism in this species. In contrast, there was evidence for significant metabolism of DCM in the mouse at high dose levels by the glutathione pathway, which leads to C02. Comparison of expired C02 levels after 4,000-ppa exposure for 6 h showed almost an ------- Toxicologies! Data 39 order of magnitude more C02 produced per kilogram of body weight in the mouse than in the rat. A marked difference was seen in the rate of clearance of DCM from tissues, as measured by its elimination in expired air. Although cleared from blood rapidly in both species, the rate of clearance from rat tissues was markedly slower than in the mouse. This slow release in the rat sustained metabolism for up to 8 h after the end of exposure and, consequently, had a marked effect on the overall body burden of metabolites. DCM was cleared from tissues in the mouse in less than 2 h. Overall, saturation of the cytochrome P-450 pathway occurred at similar levels in both species, but significantly more DCM was metabolized by the glutathione pathway in the mouse when assessed either from the blood levels of DCM or by C02 formation at high dose levels. Two physiologically based pharmacokinetic models have been formulated to predict the disposition of DCM and its metabolites. A model developed by Andersen et al. (1987) is based on inhalation exposures, whereas an alternative model developed by Angelo and Pritchard (1984) is based on intravenous and oral exposures. The latter model is discussed in detail in Sect. 4.2.3.2. Physiologically based pharmacokinetic models differ from conventional compartmental models in that they are based to a large extent on the actual physiology of the species under investigation (Clewell and Andersen 1985). Instead of using compartments based on experimentally derived time-course data, physiologically based models utilize compartments that accurately represent organ and tissue volumes, blood flow rates, and tissue partition coefficients. A model derived from this approach can qualitatively predict the time course of xenoblotic metabolism without being based directly on experimentally derived time-course data. The result is a model that predicts quantitative interspecies differences in xenobiotic absorption, distribution, metabolism, and elimination. Such models can be used to describe intraspecies and interspecies differences resulting from compound administration by different routes of exposure. Eventually, as the model is further refined by incorporating additional compartments and processes and by further iterating parameters, it becomes possible to perform quantitative extrapolations well outside the doses used in experimental studies. Ramsey, and Andersen (1984) developed a physiological pharmacokinetic model for examining the kinetic behavior of inhaled vapors and gases that are essentially nonirritating to the respiratory tract. Andersen et al. (1987) modified this model to study the metabolic disposition of DCM. The model consists of a series of simultaneous mass-balance differential equations that quantify the rate of change of the DCM concentration within tissues of the body over a specified period of time. The tissues are grouped into five compartments representing the lung, fat, liver, richly perfused organs, and slowly perfused organs. This model differs from the model described by Ramsey and Andersen (1984) because of the addition of a distinct, metabolically active lung compartment that is located between a gas exchange compartment and the systemic arterial blood. The model assumes that the inhaled air in the lung and pulmonary blood quickly achieves and maintains steady-state conditions throughout the course of exposure. None of the other organs are assumed to be in steady state. In this model, DCM enters the body through inhalation, with absorption into the pulmonary blood via gas ------- 40 Seccion 4 exchange in the lung compartment. Metabolism then occurs by two pathways in both the lung and liver compartments. The MFO pathway, which is saturable, is described by a Michaelis-Menten type equation. Metabolism by the GST pathway, which has not been demonstrated to be saturable (Gargas et al. 1986), is described as a linear, first-order process. Andersen et al. (1987) used the pharmacokinetic model to provide quantitative descriptions of the rates of metabolism by the two pathways and the levels of DCM in various organs of four mammaliam species (rats, mice, hamsters, and humans). The model, which incorporates a variety of variables representing the blood and tissue concentrations of DCM, exhaled DCM, and instantaneous rates of metabolism by each pathway, was validated by comparing predictions of DCM blood concentration and time- course data with experimentally derived results obtained with F344 racs, Syrian Golden hamsters and B6C3F1 mice, and human volunteers. The predicted values for each of the four species were in agreement with the experimental data. The model was also shown to predict reasonably well the appearance and elimination of DCM metabolites. Using this model, Andersen et al. (1987) estimated target tissue doses of DCM and its metabolites in the two bioassays conducted with the B6C3F1 mouse. In the inhalation study conducted by NTP (1986). significant increases in lung and liver tumors had been observed. In contrast, the drinking water study had failed to show a dose-related increase in the incidence of either type of tumor in the same strain of mouse. Six different dose surrogates were estimated for the B6C3F1 mouse under the conditions of each study: • Area under the liver concentration/time curve • Area under the lung concentration/time curve • Virtual concentration of metabolites derived from the MFO pathway in liver • Virtual concentration of metabolites derived from the MFO pathway in lung • Virtual concentration of metabolites derived from the GST pathway in liver • Virtual concentration of metabolites derived from the GST pathway in lung The lasc four dose surrogates represent the expected tissue exposures of several reactive metabolites that are too short-lived to measure directly (Andersen et al. 1987). They are based on the assumption that the tissue dose of a toxic metabolite is proportional to the integral of the rate of formation of the metabolite divided by tissue volume. Table 4.1 summarizes the dose surrogates estimated by Andersen et al. (1987) for the chronic bioassays performed by NTP (1986) and Serota et al. (1984). Dose surrogates related to the MFO pathway were nearly identical in the two studies. However, significant differences were observed in the dose surrogates related to the activity of the GST pathway in the inhalation and oral studies. Tumor incidence in B6C3FI mice observed in the NTP (1986) bioassay correlated with the tissue area under the concentration curve (AUC) and ------- Toxicological Daca Table 4.1. Comparison of average daily values of dose surrogates in lung and liver tissue of female B6C3F( mice in two chronic bioassays" Parameter Dose surrogate related to MFO activity in the liver Dose surrogate related to GST activity in the liver Dose surrogate related to the concentration of DCM in the liver Dose surrogate related to MFO activity in the lung Dose surrogate related to GST activity in the lung Dose surrogate related to the concentration of DCM in the lung Inhalation 2,000 3,575.0* 851.0* 362.0* 1,531.0* 123.0* 381.0" (ppm) 4,000 3,701.0 1,811.0 771.0 1,583.0 256.0 794.0 Drinking water (250 mg/kg/day) 5,197.0 15.1 6.4 1,227.0 1.0 3.1 "The chronic bioassays were performed by NTP 1986 and Serota et al. 1984. *Units are milligrams of DCM metabolized per liter of tissue. 'Units are (rag/liter)-h. Source: Adapted from Andersen et al. 1987. ------- 42 Seccion 4 che amount of DCM metabolized by the GST pathway. A similar correlation was not observed with the MFO pathway. The MFO pathway is saturable, but the GST pathway is not saturable at dose levels up to 10,000 ppm of DCM. The levels of GST-mediated metabolites increase disproportionately at high concentrations of DCM. The model predicts that in the NTP (1986) study, the values of the liver dose surrogate related to GST activity are 851 and 1,811 mg/L per 24 h after a 6-h exposure to 2,000 and 4,000 ppm of DCM, respectively. In contrast, from the drinking water study (Serota et al. 1984), the liver dose surrogate was predicted to be only 15.1 mg/L per 24 h at a dose level of 250 mgAg/day. Similarly, the values of the lung dose surrogate are 123 and 256 mg/L per 24 h after 2,000 and 4,000 ppm, respectively, whereas in the drinking water study, the dose surrogate is only 1.0 mg/L per 24 h at a dose level of 250 mgAg/day. Significant differences were also observed in the dose surrogates related to concentrations of DCM in the target tissues. The model predicted that in the NTP bioassay, the dose surrogate values related to the concentration of DCM in the liver were 362 and 771 (mg-h)/L after 2,000 and 4,000 ppm, respectively, and the values for the dose surrogate related to DCM in the lung were 381 and 794 (mg-h)/L. In contrast, the values for these parameters were 6.4 and 3.1 (mg-h)/L at a dose level of 250 mgAg/day in the drinking water study. These findings suggest that the tumor incidences observed in the two studies are not due to reactive intermediates from the MFO pathway. The estimated levels of these metabolites were similar in cases where tumors appeared and in others where they did not appear. The results are consistent with the hypothesis that (1) tumorigenicity is related to the production of reactive intermediates by the GST pathway, or (2) tumorigenicity is related to the presence of DCM in the target organs. 4.2.4.2 Oral Human. No metabolism studies data were found in the available literature on the oral administration of DCM in humans. Animal. Angelo and Pritchard (1984) developed a physiologically based pharmacokinetic model describing the disposition of DCM following exposure by intravenous dosing and by gavage. This model differs from the model of Andersen et al. (1987) in that it divides most of the organs into two subcompartments, a flow-limited vascular region and a membrane diffusion-limited extravascular region. The model includes compartments for venous and arterial blood pools, major organs (liver, kidney, lung), the GI tract, and the carcass tissue representing a large fraction of the distribution volume of DCM in the body. Simulation studies conducted by Angelo and Pritchard (1984) on dose-vehicle effects on metabolism indicate that corn oil used as the vehicle for administering DCM can affect both the uptake and distribution of DCM to target tissues and can also affect metabolism. In subsequent studies with the B6C3F1 mouse (Angelo et al. 1986a) and with the F344 rat (Angelo et al. 1986b), it was found that the route of exposure and the composition of the dosing solution had a significant effect on the pharmacokinetics of DCM. The metabolism of DCM by mice given single intravenous doses of 10 or 50 mg of 14C-DCM was ------- Toxicologies! Data 43 characterized by dose-dependent biotransformatlon to C02 and CO and rapid pulmonary clearance of the unchanged parent compound. Elimination rate constants were estimated to be 0.156/min (half-life - 4.43 min) for the 10-mg dose and 0.116/min (half-life - 5.98 min) for the 50 mg-dose. These parameters were significantly different (P < 0.05), indicating dose-dependent elimination. When DCM was administered orally to mice in single gavage doses for 14 days at levels of 50 mg/kg/day in water and 500 or 1,000 mg/kg/day in corn oil, rapid absorption and elimination were observed in the water vehicle group, whereas distinctly slower trends were observed in the corn oil vehicle group (Angelo et al. 1986a). The route and level of exposure were also found to have a significant effect on the disposition of DCM in the rat (Angelo et al. 1986b). Using the two-compartment model, Angelo et al. (1986a) estimated that the beta elimination rate constants were 0.058/min (half-life - 11.9 min) in rats receiving a single intravenous dose of 10 mg and 0.028/min (half-life - 23.5 min) in rats administered a single 50-mg intravenous dose of DCM. The disposition rate constants were significantly different (P < 0.05), indicating dose-dependent elimination from the blood. The metabolism of DCM was also found to be dose dependent. When DCM was administered to rats by gavage at daily doses of 50 or 200 mg/kg/day for 14 days, rapid absorption and distribution to the tissues were observed. Dose-dependent biotransformation of DCM to C02 and CO and rapid pulmonary clearance of the unchanged parent compound characterized the disposition of DCM. McKenna and Zempel (1981) reported that in rats (250 g) given 1 or 50 mg/kg of l^C-DCM, -5% or 2% of the dose, respectively, was excreted in the urine within 48 h. Mice given DCM orally in corn oil at a dose of 100 mg/kg eliminated 40% of the dose in the expired air as unchanged DCM, 20% as CO, and 25% as C02 within 96 h; a dose of 1 mg/kg DCM was metabolized to CO (45%) and C02 (50%) (Yesiar et al. 1977). 4.2.4.3 Dermal Human: No metabolism studies were found in the available literature on the dermal administration of DCM. Animal. McDougal et al. (1986) developed a physiological model for the penetration of organic vapors through skin in vivo. The model can be used to protect blood concentrations (after dermal vapor exposures in the rat) when chemical distribution coefficients, physiological and metabolic parameters, and skin permeability constants are known. Permeability constants for DCM were calculated by using a physiologically based pharmacokinetic model for dihalomethanes (Gargas et al. 1986) to relate blood concentrations during dermal vapor exposures to the total amount of chemical that was absorbed through the skin. A skin compartment was added to the model which had input based on the permeability-area concentration product. This predictive model adequately described blood concentrations after DCM dermal vapor exposures over a wide range of doses. ------- 44 Section 4 4.2.4.4 In vitro studies In vitro studies (Anders et al. 1977, Gargas et al. 1986) demonstrated that the metabolism of DCM involved two pathways (Fig. 4 1) Gargas et al. (1986), cited in EPA 1987a. predicted that the microsomal metabolism of DCM might yield both CO and C02- The mechanism prediccs that following the initial oxidation of DCM to formyl chloride, this intermediate combines with glutathione and is then further metabolized by a GST to C02. Gargas et al. (1986), cited in EPA 1987a, further suggested that the formation of the formyl glutathione intermediate is possible, assuming that formyl chloride is a relatively stable intermediate. The metabolism of DCM was compared in vitro using rat, mouse, and hamster lung and liver fractions (CEFIC 1986a). Comparisons were also made between metabolism in animal tissues and that in human liver in vitro. Tissue homogenates were separated into microsomal and cytosolic fractions to investigate the cytochrome P-450 and glutathione-S- transferase dependent pathways of DCM metabolism, respectively. The cytochrome P-450 pathway was assessed by measuring the conversion of DCM to CO, and the glutathione-S-transferase pathway was determined by measuring formaldehyde formation. Metabolic rates were determined ac a series of substrate concentrations, and, where possible, the metabolic rate constants Km and Vmax were obtained. The most active tissue for metabolizing DCM by the cytochrome P-450 pathway was the hamster liver, with a similar rate observed in mouse liver. The rates for rat and human liver were similar but significantly lower than those found in the mouse and hamster. A parallel difference in response was seen in lung tissue, with rat lung showing significantly lower metabolic rates than the mouse and hamscer lung. Human lung tissue was not available. For the glutathione-S-transferase mediated pathway, mouse liver was considerably more active than any other tissue, the maximal rate being more than 12-fold greater than the next most active tissue, the rat liver. No activity could be detected for this pathway in either hamster or human liver, even though these tissues were active with another substrate for the glutathione-S-transferases. A low rate of metabolism of DCM by this pathway could be detected in mouse lung but not in rat or hamster lung. Metabolism by the glutathione pathway correlates well wich the known carcinogenicity of DCM in animals, whereas metabolism by the cytochrome P-450 pathway does not. 4.2.5 Excretion 4.2.5.1 Inhalation Human. The urinary excretion of DCM was measured in 11 human subjects after inhalation exposure (DiVincenzo et al. 1972). Less than 2% was detected in urine unchanged. In four subjects exposed to 100 ppm (347 mg/m3) DCM (purity not specified) for 2 h, an average of 22.6 ng DCM was excreted in the urine within 24 h after the exposure; in seven ------- CYT P-450 CYTOSOL Oj> s\ iA NADPH i GSH X JH2-X ),<- II H'- i ^X <-— >— - NUCLEOPHILE? (i.e.. GSH) b ^H* GS-CH2 -OH CH20 + GSH NAD* -V° I XH C02 HCOOH * GSH I I C02 H GS-C H GSH i CO2 n o o B) n Di Fig. 4.1. Proposed pathways for melhylenc chloride metabolism. ------- 46 Section 4 subjects exposed to 200 ppm (694 mg/m3) DCM for 2 h, the corresponding value was 85.5 /ig. No data were found on amounts recovered In feces or urine. Animals. No excretion studies were found in the available literature on the inhalation exposure of DCM in humans. 4.2.5.2 Oral Human. No excretion studies were found in the available literature on the oral administration of DCM in humans. Animal. In rats, a single oral dose, or 1 or 50 mg/kg 14C-DCM, was eliminated in expired air as unchanged DCM (12.3 or 72%, respectively) within 48 h. For corresponding duration and dose, fecal (<1%) and urinary (5%, 2%, or the administered dose, respectively) eliminations were low (McKenna and Zempel 1981). 4.2.5.3 Dermal No excretion studies were found in the available literature on the dermal administration of DCM in humans. Animal. No excretion studies were found in the available literature on the dermal administration of DCM in animals. 4.2.6 Discussion The current evidence is not sufficient to identify with reasonable certainty the carcinogenic mechanism of action of DCM. The available data suggest that DCM produces a carcinogenic response via GST-mediated metabolism. There is a strong correlation between GST activity and the occurrence of tumors in mice. In addition, there is little evidence to implicate either parent DCM or metabolites resulting from the alternative MFO pathway. The model developed by Andersen et al. (1987) provides a framework for estimating the nonlinearity inherent in the metabolism of DCM by two pathways used to different extents at high and low doses in the risk estimation procedure. When applied to high-Co-low dose extrapolation, the model is relatively Insensitive to major uncertainties in the pharmacokinetic data. It accounts for the nonlinearities in the internal doses across exposure levels that arise from dose-dependent changes in absorption, distribution, excretion, and saturation of metabolism. It characterizes the nonlinearity of metabolism via the GST pathway at low and high doses as a result of saturation of the competing MFO pathway. This model can be used to extrapolate to human DCM vapor exposure conditions by substituting human physiological parameters for the animal values. Andersen et al. (1987) used their model to estimate target tissue doses of presumed toxic chemical species in humans exposed to DCM by inhalation or by drinking water. Estimated target tissue doses in humans exposed to low concentrations of DCM were several fold lower than linear extrapolation from high-dose mice would predict. The EPA (1987a,b) has pending an approximate ninefold reduction in its inhalation unit risk, taking into account the model's results. Andersen et al. (1987) advocate a much larger reduction, from 140 to 210 fold, based on a different ------- lexicological Data 47 interpretation of EPA's surface area factor for interspecies extrapolation Questions about interspecies extrapolation, using the model, result from unresolved issues about the impact of differences in species sensitivity to a given internal, target-tissue dose. 4. 3 TOXIC ITY 4.3.1 Overview The toxic effects of DCM are varied. Methylene chloride may cause acute lethality when animals are administered large doses orally, intraperitoneally, subcutaneously, or by inhalation. Fatty degenerative changes, glycogen depletion in the liver, and kidney damage were seen after inhalation exposure to large doses of DCM. Methylene chloride was toxic to the cardiovascular system at high doses. Decreased blood pressure and increased heart rate were reported in primates. Central nervous system effects included behavioral and performance deficits, depression, anesthesia, coma, and death. Chronic oral exposure to DCM via drinking water resulted in hepatic histopathological alterations, decreased body weight gain, and biochemical changes in the blood of rats. In animals, no prominent teratogenic potential has been shown to exist for DCM. Results were negative in a two-generation reproduction study. Mutagenic activity has been observed in some strains of bacteria and yeast and in a mammalian cell culture line. Results were largely negative in mammalian cells in vivo. Chronic exposure to DCM in drinking water produced a significant increase in liver tumors in female rats when compared with matched controls; however, the tumor incidences were within historical control levels. No carcinogenic response was indicated in mice. An EPA assessment of the results of these studies suggested a borderline statistical increase in the incidence of hepatic neoplastic nodules and carcinomas (combined) in female Fischer 344 rats and male B6C3F1 mice. Sarcomas have been reported in the salivary gland region of male rats; however, the authors suggested that a viral disease in rats may have contributed to the tumorigenic response. The NTP reported clear evidence of the carcinogenicity of DCM in female F344/N rats and in B6C3F1 mice of both sexes in a chronic inhalation carcinogenicity bioassay. Methylene chloride induced increased incidences of benign neoplasms in the mammary gland in rats and of alveolar/bronchiolar neoplasms and hepatocellular neoplasms in mice of both sexes. 4.3.2 Lethality and Decreased Longevity 4.3.2.1 Overview Exposure to DCM can be fatal in humans and animals following inhalation and ingestion. Lethal dose levels could not be determined in humans; however, LCSO values of 11,000 to 16,000 ppm were reported. Oral exposures were lethal at doses of 1,000 to 4,000 mg/kg. 4.3.2.2 Inhalation Human. Case studies of DCM poisoning have demonstrated that exposure can be fatal. One case of accidental death resulted from acute DCM exposure during paint-stripping operations (Bonventre et al. 1977). Methylene chloride concentration was not reported; however, it was detected in various tissues, including the liver (14.4 mg/100 g of ------- 48 Section 4 tissue), blood (510 mg/L), and brain (24.8 mg/100 g of tissue). Another death occurred following acute exposure to DCM being used as a paint remover (Stewart and Hake 1976). In this case, no data were provided on DCM concentrations or on the tissue levels of DCM detected. No lethality data were found in the available literature following chronic exposure to DCM. Animal. When laboratory animals inhaled DCM, guinea pigs were more susceptible than mice to its lethal effects (Table 4.2). 4.3.2.3 Oral Human. No lethality studies were found in the available literature on the oral administration of DCM in humans. Animal. When exposed orally, CF-1 mice and Sprague-Dawley rats were about equally susceptible to the lethal effects of DCM (Table 4.2). 4.3.2.4 Dermal Human. No studies were found in the available literature on the lethal effects of dermally applied DCM in humans. Animal. No studies were found in the available literature on the lethal effects of dermally applied DCM in animals. 4.3.2.5 Discussion Two human case studies have demonstrated that DCM exposure can be fatal. Methylene chloride was lethal when inhaled by animals at high concentrations. LC50 values of 11,000 to 16,000 ppm were reported. Similar responses were noted at 1,000 to 4,000 mg/kg following oral administration. Based on animal studies, it appears that high levels are necessary to cause mortality. 4.3.3 Systemic/Target Organ Toxicity 4.3.3.1 Overview Various human and animal studies demonstrated that the liver and the central nervous system (CNS) are the primary targets following DCM exposure. The primary route of entry to the body for DCM is via inhalation, and most of the studies found in the available literature are concerned with inhalation. In acute experimental inhalation studies in humans, DCM administered at concentrations in excess of 300 ppm for up to 4 h altered behavioral performance, as manifested by decreased visual and auditory functions. Similarly, various psychomotor tasks were impaired, but these occurred at higher levels (800 ppm). Acute animal studies support findings of DCM-induced effects in the central nervous system. These effects occurred, however, at higher concentrations for corresponding durations of exposure. Slight narcosis occurred in several species at 4,000 to 6,000 ppm (2.5 to 6 h). Behavioral effects, as manifested by the reduction in rapid eye movement during sleep, were also demonstrated after 24 h (1,000 to 3,000 ppm). ------- TOXLCOlogical Data Table 4.2. Acute lethality of methylene chloride Species (strain/sex/No.)fl Mouse (CF-l/M/50) Mouse (S.P.F, OF,/F/not stated) Mouse (Swiss/not stated/80) Guinea pig ( Hartley /M/60) Mouse (CF-l/M/65) Rat (Wistar/M/not stated) Rat (Sprague-Dawley/ M/not stated) Route (duration) Inhalation (20 min) Inhalation (6h) Inhalation (8h) Inhalation (6h) Oral Oral Oral Effect Dose* LC$o 26,710 ppm (92,649 mg/m3) LCso 14.155 ppm (49,1 18 mg/m3) LCso 16,189 ppm (56, 176 mg/m3) LCso 11, 600 ppm (40,252 mg/m3) LDjo 1,987 mg/kg LD95 5.16mM/100g (4.382 mg/kg) LDso 1.6 mL/kg (2,121 mg/kg) References Aviado et al. 1977 Gradiski et al. 1978 Svirbely et al. 1947 Balmer et al. 1976 Aviado et al. 1977 Ugazio et al. 1973 Kimura et al. 1971 "M = male, F •* female. No. =" total number of animals in lethality study. * Expressed as exposure for inhalation route. Source: Adapted from EPA 1985c. ------- 50 Section 4 Various inhalation and oral animal studies revealed mild liver effects following DCM exposure. Alterations in liver cytochromes were demonstrated in animal species following inhalation of 500 ppm DCM for 10 days. Cellular changes were observed at 100 ppm (100 days). In oral studies, histomorphological alterations were reported in animals exposed to 250 mg/kg/day OCM for 104 weeks. Limited data show that high concentrations of DCM can cause upper respiratory tract irritation and produce cardiovascular, ocular, and renal effects. 4.3.3.2 Central nervous system Central nervous system effects have been observed in humans in experimental studies following DCM exposure. Alterations in behavioral performance and various psychomotor tasks were evident after short-term exposure. Neurobehavioral effects were reported in factory workers exposed to DCM. Similarly, CNS effects have been demonstrated in animals following acute exposure, but these occurred at exceedingly high dose levels. Inhalation, human. A primary adverse health effect associated with short-term exposure to DCM is impairment of CNS function (Table 4.3). In experimental studies, inhalation of DCM for up to 5 h decreased visual and auditory functions (>300 ppm) and various psychomotor tasks (800 ppm) (Fodor and Winneke 1971, Winneke 1974). Similarly, CNS effects were produced at higher dose levels and for short exposure periods as alterations in visually evoked responses were detected 1 to 2 h following exposure to 1,000 ppm (Stewart et al. 1972). Longer-term exposure also produced CNS effects (Table 4.4). Neurotoxicity was the most prominent symptom complex reported in over 100 cases involving occupational exposure to DCM. Welch (1987) reported that workers from several industries, including auto parts production and plastic and prosthesis manufacturing, presented a variety of CNS complaints, including headaches, dizziness, nausea, memory loss, parasthesia, tingling in hands and feet, and loss of consciousness. These effects occurred during certain painting, cleaning, and spraying operations:"Methylene chloride levels measured were up to 100 ppm, and duration of exposure was 6 months to 2 years. Levels greater than 100 ppm (duration not specified) produced corresponding effects under similar situations. It should be noted that workers were exposed concomitantly to other unspecified solvents. Results of this study should be viewed cautiously. In another study, subjective assessments of sleepiness, physical tiredness, and mental tiredness were evaluated in factory workers (Cherry et al. 1983). A total of 56 workers were exposed to 28 to 173 ppm of DCM (in a 9:1 DCM/methanol atmosphere). The changes in parameters measured were significantly greater (P < 0.05 for mental tiredness, P < 0.01 for physical tiredness and sleepiness) only for the morning shift, and the magnitude of the changes correlated in a statistically significant manner with the blood CO-Hb levels at the end of the shift Although no statistically significant changes were observed in two objective tests (digit symbol substitution test and reaction time), deterioration in performance on the morning shift correlated (P < 0.01) with the end-of-shift blood concentration of DCM. ------- Toxicological Data 51 Table 4.3. Short-term experimental metbylene chloride inhalation exposures and reported effects in humans Concentration (ppm) Duration Effects References 500-1,000 1-7.5 h/day CO-Hb percentages proportional 5 days/week to exposure concentration and time; CNS depression at 1000 ppm 50-500 7.5 h Decreased dissociation of oxygen 5 days/week from Hgb in proportion to exposure concentration 100 and 500 7.5 h/day Blood lactic acid increased 5 days/week slightly from exercise at 500 ppm, not 100 ppm 317 and 751 4 h Depressed CFF," decreased auditory vigilance performance 317, 470, 3-5 h Decreased performance of CFF," 751 auditory vigilance, and psychomotor tasks "Critical Fucher Frequency. Stewart et al. 1972 Forster et al. 1974, cited in NIOSH 1976 Fodor and Winneke 1971 Winneke 1974 ------- 52 Section 6 Table 4.4. Longer-term occupational methylene chloride inhalation exposures and effects in humans Concentration (ppm) Duration Effects References Weiss 1967. cited in NIOSH 1976 660-3,600 One worker. After 3 years: burning pain several around heart, restlessness, hours per day feeling of pressure, for 5 years palpitations, forgetfulness, insomnia, feeling of drunkeness. After 5 years: auditory and visual hallucinations, slight erythema of hands and underarms. Diagnosed as having toxic encephalosis. 28-173 Reversible subjective symptoms Cherry et al. 1983 ------- Toxicologies! Data 53 Inhalation, animal. Short-term inhalation studies showed that DCM produced central nervous system effects (Table 4.5). In several species. slight narcosis occurred at 6000 ppm after 2.5 h exposure (Ueinstein et al. 1972); deep narcosis occurred in rats at 16,000 to 18,000 ppm after 6 h exposure (Berger and Fodor 1968). Behavioral effects have also been reported. A reduction in paradoxical sleep was observed in rats exposed at 1,000 or 3,000 ppm for 24 h. This effect on sleep was not observed at 500 ppm (Fodor and Uinneke 1971). Inhalation of 500 ppm for 6 h daily for 4 days resulted in an increase in preening frequency in rats (Savoleinen et al. 1977). In studies using the running-wheel technique, there was decreased spontaneous motor activity in male rats exposed intermittently to 500 ppm DCM for 7 h/day, 5 days/week for up to 6 months (Heppel et al. 1944, Heppel and Neal 1944). An inhalation neurotoxicity study on DCM in rats exposed to 50, 200, and 2,000 ppm DCM for 90 days is currently under way. This study is sponsored by Halogenated Solvents Industry Alliance (HSIA 1988) and includes electrophysiology, a functional observational battery, and neuropathology as end points. HSIA (1988) reported that preliminary results show that DCM did not produce neurotoxic effects upon repeated exposure in rats. Results of this study are expected to be available in the spring of 1988. Oral, human. No CNS or behavioral studies were found in the available literature on the oral administration of DCM in humans. Oral, animal. No CNS or behavioral studies were found in the available literature on the oral administration of DCM in animals. Dermal, human. No CNS behavioral studies were found in the available literature on the dermal administration of DCM in humans. Dermal, animal. No CNS or behavioral studies were found in the available literature on the dermal administration of DCM in animals. Discussion. Based on experimental data, it appears that impairment of behavioral or sensory responses may occur in humans following acute inhalation exposure to DCM at levels exceeding 300 ppm for short duration, and the effects are transient. No data were found in the available literature on CNS effects after oral or dermal exposure. The CNS effects produced following exposure to DCM are probably due to DCM alone or in combination with CO-Hb. Evidence has been provided which shows that the MFO pr.thway (resulting in CO production) is mostly saturable, producing maximum blood CO-Hb levels of <9%. These CO-Hb levels are not sufficiently high enough to induce the CNS effects observed following acute DCM exposure; therefore, it appears that behavioral effects are probably due to DCM alone or in combination with CO-Hb. 4.3.3.3 Hepatotoxicity Available literature demonstrated that DCM can adversely affect the liver in animals, and suggestive evidence for a similar effect in humans was also found. In animal inhalation studies, cellular changes were ------- 54 Section Table 4.3. Short-tern exposures ud effects la Concentration (ppm) 14.500 Duration 2b Effects Mice — death References Flury and Zermk 1931 10.000 4.000 6.000 15.000 and 20.000 40.000 2h 6h 6h 23.000-28.000 16.000-18.000 lib 6h 5.000-9.000 2.800 3.000 and 1.000 5,000 5.000 8h 14 h 24 b 30min/ day 7dayi Mice — narcosis Dogs — light narcosis after 2 5 h. rabbits after 6 h Guinea pigs— slight narcosis in 2 5 h. rabbits and cats in 3-4 h, dogs in 2 h Dogs — loss of pupillary and corneal reflexes after 10-20 mm: complete muscular relaxation after 25-35 mui; reduction in blood pressure; rapid narcosis at 20.000 ppm Dogs — loss of pupillary and corneal reflexes after 10-20 mm; complete muscular relaxation after 16 min: 3 of 5 dogs died from progressive hean failure due to cardiac injury Rats— electrical activity stopped after 1.5 h Rats— initial excitement followed by deep narcosis, decreased EMG tonus; decreased EEC activity; breathing difficulties: tremor, electrical activity stopped after 6 h Long sleeping phase lacking desynchronuation phase* Rats— de d proportion of REM sleep to total sleep Rats—suppressed REM sleep: increased tune between two REM periods; linear relation between dose and response Rats—decreased running activity Mice—initial increase in physical activity followed by decrease in food and water intake, lethargy, increased liver to body weight ratio and liver fat. mild fatty infiltrations, hydropic degeneration of centnlobular cells Wemstem et al. 1972 Von Oettingen et al. 1949 Berger and Fodor 1968 Fodor and Winneke 1971 Heppel and Neal 1944 Weinstein et al. 1972 ------- Tabfe4.5 (condoMd) Toxicological Data 55 Concentration (ppm) Duration Effects References 5.200 500 5.000 4,500 1.2SO 6 h Fatty infiltration of liver Moms et al. in guinea pigs. Effects were 1979 transient in mice 10 days Altered cytochromes P-450: Norpotb et al. reduction in ammopynne 1974 /V-demethylase activity in rats 10 days Elevation of enzyme activity Norpoth et al. 1974 10 days Increased maternal absolute Hardin and and relative liver weight Maason 1980 and lowered fetal weight* in rats 10 days Lowered fetal weights and Schwetz et al. increased maternal average 197S absolute liver weight in rats Increased maternal liver weights Source. Adapted from NIOSH 1976. EPA 1985a. I985c. ------- 56 Section 4 demonstrated at 100 ppm (100 days), and liver cytochromes were altered following a 10-day exposure at 500 ppm. It should be noted that hepatic effects reported at 100 ppm were elicited upon continuous exposure without recovery as opposed to the more traditional exposure protocol. An increased incidence of hepatocellular vacuolization was observed in male and female rats exposed to 500 ppm. Female rats exposed to 500 ppm DCM also had an increased incidence of multinucleated hepatocytes (Nitschke et al. 1982. 1988). In oral studies, histomorphological alterations were observed in animals exposed to 50 mg/kg/day DCM for 104 weeks. Inhalation, human. Welch (1987) reported a case of hepatitis in a worker who was exposed to solvents. Methylene chloride was used in combination with other unspecified solvents, but the DCM levels measured in the serum were significantly higher than for the other solvents. There were no exposure measurements reported for DCM or other solvents. Several workers in similar work environments had abnormal liver function tests, but no measurements of exposure were reported. Results from this study should be viewed cautiously. Inhalation, animal. Acute and chronic studies have revealed the liver as a target organ following DCM exposure (Tables 4.5 and 4.6). Acute studies showed that DCM altered liver structure and cytochrome activity (Table 4.5). The inhalation of 5,200 ppm for 6 h caused fatty infiltration in the liver of guinea pigs (Morris et al. 1979); transient fatty changes were also observed in mice exposed to 5,000 ppm for 7 days (Ueinstein et al. 1972). Following a 10-day exposure to vapors of DCM, a significant increase in liver cytochrome P-450 occurred in male SPF Wistar rats exposed to 500 ppm. The increase did not occur in animals exposed to 5,000 ppm (Norpoth et al. 1974). No statistically significant change occurred at the high dose. The author reported that the inverse dose-response relationship for P-450 may be due to the combined effect of DCM and CO. Differential enzymatic induction was also reported. Reduced liver enzyme (aminopyrine N- demethylase) activity occurred in animals exposed to 500 ppm; however, activity was elevated in those animals exposed to 5,000 ppm (Norpoth et al. 1974). After 28 days of exposure to DCM (250 ppm), no changes were noted in liver enzymes (Veinstein et al. 1972). Liver toxicity was also observed following longer-term exposures by inhalation (Table 4.6). Continuous inhalation exposure to DCM (100 ppm) for 10 weeks resulted in centrilobular fat accumulation in ICR mice. The increase in fat was accompanied by a decrease in glycogen that persisted with termination of the study at 10 weeks (Ueinstein and Diamond 1972. cited in EPA 1985c). Cytoplasmic vacuolization and positive fat staining were reported in mice exposed continuously via inhalation for 100 days to 100 ppm DCM. Cytochrome P-450 was decreased but P-b5 was increased (Haun et al. 1972). The Haun study also reported cytoplasmic vacuolation and the presence of fat droplets in the liver of rats and dogs exposed to 25 to 100 ppm for 100 days, and fatty degeneration in the liver of monkeys exposed to 1,000 ppm for 100 days. However, the liver effects reported in this study were elicited upon continuous exposure. Male and female Sprague-Dawley rats exposed to DCM (0, 50, 200, or 500 ppm) for 2 years showed an increased incidence of multinucleated hepatocytes at 500 ppm (Nitschke et al. 1982). Exposure to DCM (1,000 ppm-low dose. ------- Toxicological Daca 57 Table 4.6. Longer-term methyleoe chloride inhalation exposures and effects in animals Concentration (ppm) 5.000 10.000 1,000 5,000 100 25 and 100 25 and 100 1,000 ppm 100 25 100 500 50, 123. 250*'b 500. 1.500. or 3.500 1.000 or 4,000 Duration 7 h/day. 5 days/week up to 6 months 4 h/day. 5 days/week. 18 weeks 14 weeks continuous 14 weeks continuous 100 days 100 days 100 days 100 days 90 days 14 weeks continuous 10 weeki continuous 2 yean 2 yean 2 yean 2 yean Effects Various experimental animals — no effect Various animals — mcoordination. conjunctiva! irritation: shallow respiration, pulmonary congestion, edema with focal extravasation of blood, some fatty degeneration Various experimental animals — increased hematocnt, Hgb. RBC, bihrubin. weight loss: mild centnlobular fat High mortality: pneumonia: fatty liver, icterus: splenic atrophy; edema of meninges; renal tubule vascular changes Mice — liver cytoplasmic vacuolization and positive fat staining Rats — nonspecific renal tubular degenerative and regenerative changes Dogs — mild cytoplasmic vacuolation and sinusoidal congestion in the liver Rhesus monkeys— centnlobular fatty degeneration of the liver Mice — hepatic cytochrome P-450 decreased and bj increased Mice — increased activity Elevated liver fat: decreased hepatocyte glycogen: centnlobular fatty infiltration Rats — multmucleated hepatocytea Rats — hepatocellular alteration and fatty infiltration Rau — multinucleated hepatocytes: liver cytoplasmic vacuolization Rats — increased hemosiderosu; cytomegaly and cytoplasmic vacuolization in the liver References Heppel et al 1944 MacEwen et al. 1972 Haun et ai. 1972 Haun et al. 1972 Haun et al 1972 Thomas et al. 1972 Wemstein and Diamond 1972 Nitschke et al. 1982, 1988 NCA 1982 Burek et al. 1984 NTP 1986 •mg/kg/day. Route of exposure-drinking water Source NIOSH 1976: EPA I985a.c. ------- 58 Section 4 4.000 ppm-high dose) was associated with Che increased incidence of nemosiderosis, cytomegaly, and cytoplasmic vacuolizacion (NTP 1986). Oral, human. No hepatotoxlcity studies were found in the available literature on the oral administration of DCM in humans. Oral, animal. Liver toxicity was also observed following long-tern exposures by oral administration (Table 4.5). Increased incidence of foci and areas of cellular alteration were detected in Fischer 344 rats administered 50 to 250 mg/kg/day DCM in drinking water for 2 years (NCA 1982). Also, fatty liver changes were detected in the 125- and 250- ragAg/day dose groups at 78 and 104 weeks of treatment. No liver effects were observed at 5 mg/kg/day (NCA 1982). Dermal, human. No hepatotoxicity studies were found in the available literature on the dermal administration of DCM in humans. Dermal, animal. No hepatotoxicity studies were found in the available literature on the dermal administration of DCM in animals. Discussion. Based on available data, histomorphological changes of the liver can occur following short-term inhalation exposure (6 h to 7 days) at high dose levels (5,200 ppm). Alteration in cytochrome activity can occur at lower levels (500 ppm for 10 days). Following longer-term exposure, liver effects may occur at concentrations greater than 100 ppm (100 days) following continuous inhalation exposure, and greater than 50 mg/kg/day via ingestion (for 2 years). Animal data presented liver effects that consisted primarily of histologic alterations of liver cells, such as those seen in aging animals. Further, the fatty changes reported were reversible. Liver toxicity was not reported in epidemiological studies. Therefore, it appears that DCM will not cause serious liver effects in humans at the higher levels reported in occupational settings. 4.3.3.4 Renal effects Available studies show that DCM may cause kidney effects such as congestion and tubular changes. Inhalation, human. Several epidemiological studies were conducted; however, no association was found between DCM exposure and adverse kidney effects (Friedlander et al. 1978, Ott et al. 1983a, Heame et al. 1987). Inhalation, animal. No data were found in the available literature on the renal toxicity of DCM following short-term exposure. However, continuous exposure to DCM (25 and 100 ppm) for 100 days via Inhalation in rats shoved nonspecific renal tubular degenerative and regenerative changes; no changes in organ-body weight ratios were found (Haun et al. 1972) (Table 4.5). These effects were elicited upon continuous exposure to DCM. Oral, human. No renal studies were found in the available literature on the oral administration of DCM in humans. Oral, animal. No renal studies were found in the available literature on the oral administration of DCM in animals. ------- Toxicological Data 59 Dermal, human. No renal studies were found in the available literature on the dermal administration of DCM in humans. Dermal, animal. No studies were found in the available literature on the renal effects of dermally applied DCM in animals. Discussion. The paucity of data on renal effects in humans and animals precludes any determination of the significance of this biological end point as a factor in the toxicity of DCM. 4.3.3.5 Respiratory effects Available data show that DCM may affect the respiratory system. Irritant effects were primarily reported. Inhalation, human. Exposure to DCM in the range of 100 ppm appeared to cause significant upper respiratory irritation (Velch 1987). Inhalation, animal. No studies were found in the available literature on the respiratory effects of inhalation exposure to DCM in animals. Oral, human. No studies were found in the available literature on the respiratory effects of inhalation exposure to DCM in humans. Oral, animal. No studies were found in the available literature on the respiratory effects of orally administered DCM in animals. Dermal, human. No studies were found in the available literature on the respiratory effects of dermally applied DCM in humans. Dermal, animal. No studies were found in the available literature on the respiratory effects of dermally applied DCM in animals. Discussion. The paucity of data on the respiratory effects of DCM in humans precludes any determination of the significance of this biological end point as a factor in the toxicity of DCM. 4.3.3.6 Cardiovascular Effects on the cardiovascular system were noted in animals, particularly at high dose levels. In humans, certain individuals with an existing cardiac disease may be more susceptible to the toxic effects of DCM. Inhalation, human. Ott et al. (1983b) reported on SO employees of two fiber production plants who were selected for study--24 of whom were occupationally exposed to DCM. All of the participants were white males between 37 and 63 years of age. Eleven of the men had reported a history of heart disease. Under the conditions of this study, neither an increase in ventricular or supraventricular ectopic activity nor episodic ST-segment depression was associated with exposure to DCM that ranged from a TWA of 60 to -475 ppm. Inhalation, animal. Myocardial contractility was altered following short-term exposure to DCM, usually at concentrations >20,000 ppm (Aviado and Belej 1974). No data were found on long-term exposures. ------- 60 Section 4 Oral, human. No studies were found in the available literature on the cardiovascular effects of orally administered DCM in humans. Oral, animal. No studies were found in the available literature on the cardiovascular effects of orally administered DCM in animals. Dermal, human. No studies were found in the available literature on the cardiovascular effects of dermally applied DCM in humans. Dermal, animal. No studies were found in the available literature on the cardiovascular effects of dermally applied DCM in animals. Discussion. Methylene chloride produces cardiotoxic effects; however, these effects are most likely mediated by CO via CO-Hb. Data from experimental exposure studies indicate that blood CO-Hb levels, following exposure to DCM, are elevated in a manner that is dependent on the inhaled concentration of DCM and the length of exposure. Exposure to DCM at concentrations that do not exceed the current OSHA standard of 500 ppm (1,740 mg/m3), as an 8-h TWA with a ceiling of 1,000 ppra and a maximum peak of 2,000 ppm (5 min in 2 h), can elicit CO-Hb levels of about 3 to 6%. Exposures at a higher concentration, 1,000 ppm for up to 2 h, result in CO-Hb levels of up to 10%. The reported CO-Hb concentrations of 3 to 10%, although not expected to cause adverse health effects in normal healthy individuals from occasional product use, may be of concern to certain sensitive populations such as those with compromised cardiovascular systems. Moreover, since CO is known to bind to myoglobin and disrupt metabolism, this could affect cardiac function, especially with underlying heart disease. 4.3.3.7 Ocular effects Exposure of rabbits to vapors of DCM (17,500 mg/m3) resulted in increased corneal thickness and intraocular tension (Ballantyne et al 1976). 4.3.4 Developmental Toxicity 4.3.4.1 Overview Limited studies found in the available literature suggested that when DCM was inhaled at concentrations of 1,250 ppm and above, it was developn»ntally and maternally toxic. 4.3.4.2 Inhalation No developmental toxicity studies were found in the available literature on the inhalation of DCM in humans. Animal. Studies have demonstrated that DCM crossed the placental barrier (Anders and Sunram 1982). Fetuses of Swiss-Webster mice and Sprague-Dawley rats exposed to DCM (1,250 ppm) on days 6 to 15 of gestation showed cleft palate and rotated kidneys (mice), dilated renal pelvis (rats), and delayed ossification of sternebrae (mice and rats) (Schwetz et al. 1975). These effects were not statistically significant except for a minor skeletal variant (extra sternebrae, P < 0.05). Developmental toxicity, as evident by Lowered fetal body weights, was ------- Toxicological Daca 6L reported in rats at 4,500 ppm (-15,600 mg/m3) (Hardtn and Hanson 1980) It should be noted that these effects were also observed at minimally maternally toxic levels. In rats exposed to 1,250 ppm, the average absolute maternal liver weight was significantly increased over control levels (Schwetz et al. 1975). In mice, there was a significant increase in maternal body weight and absolute liver weight at 1,250 ppm (Schwetz et al. 1975). Maternal toxicity, manifested by increased absolute and relative liver weights, was reported in rats at 4,500 ppm. Malformations in chick embryos were reported when DCM was injected into the air sacs of fertile eggs (Elovaara et al. 1979). The significance of these malformations and their relevance to mammalian teratology is unclear. 4.3.4.3 Oral Human. No studies were found in the available literature on the developmental toxicity of orally administered DCM in humans. Animal. No studies were found in the available literature on the developmental toxicity of orally administered DCM in animals. 4.3.4.4 Dermal Human. No studies were found in the available literature on the developmental toxicity of dermally administered DCM in humans. Animal. No studies were found in the available literature on the developmental toxicity of dermally administered DCM in animals. 4.3.4.5 Discussion The practical importance of these findings is limited since each of the two studies used only one dose level, and these effects were observed at a maternally toxic dose. Further, the effects on fetal development in mice were minor. The use of only one dose level precludes any evaluation or dose-response relationships. Evidence, therefore, does not currently exist to conclusively characterize the developmentally toxic potential of DCM. 4.3.5 Reproductive Toxicity 4.3.5.1 Inhalation Human. No reproductive toxicity studies were found in the available literature on the inhalation of DCM in humans. Aniaal. No adverse effects on reproductive parameters, neonatal survival, or neonatal growth were observed in F344 rats exposed to 0, 100, 500, or 1.500 ppm DCM for 6 h/day, 5 days/week for 14 weeks in either FO or Fl generations (Nitschke et al. 1985). Similarly, there were no treatment-related gross pathological effects on FQ and Fl adults and Fl and F2 weanlings at necropsy. Histopathological examination of tissues from Fl and F2 weanlings did not reveal any lesions attributed to DCM toxicity. ------- 62 Section 4 4.3.S.2 Oral Human. No studies were found In the available literature on the reproductive effects of orally administered DCM In humans. Animal. When DCM (125 ppm) was administered in the drinking water of male and female rats for 91 days, there were no effects on the estrus cycle or on reproduction (Bornmann and Loesser 1967). 4.3.5.3 Dermal Human. No studies were found In the available literature on the reproductive effects of dermally applied DCM in humans. Animal. No studies were found in the available literature on the reproductive effects of dermally applied DCM In animals. 4.3.5.4 Discussion Only one study (two-generation) was found In the available literature that evaluated end points acceptable for assessing reproductive toxicity. The results of this study were negative. Based on these data, DCM does not appear to pose a hazard to human reproduction. 4.3.6 Genotoxicity The genotoxicity of DCM has been evaluated in a variety of mammalian test systems (in vitro and in vivo) in studies by Andrae and Wolff 1983; Burek et al. 1984; CEFIC 1986c,d,e; Gocke et al. 1981; Jongen et al. 1981; McCarroll et al. 1983; Perocco and Prodi 1981; and Thilagar and Kumaroo 1983, 1984a,b. Results of these studies are summarized in Table 4.7. Methylene chloride reportedly produced a dose- related increase in chromosomal aberrations in human peripheral lymphocytes, in Chinese hamster ovary (CHO) cells, and In mouse lymphoma L5178Y cells. Methylene chloride was tested in CHO cells at 2, 5, and 10 /iL/mL in the presence and absence of an S9 system and at 15 /*L/mL in the presence of the system (Thilagar and Kumaroo 1983). Chromosome damage occurred at all dose levels, and the extent of damage was greater in the presence of activation. Maximum levels of chromosome damage were obtained in the presence of S9 at the highest DCM concentration (15 ML/mL) (Thilagar and Kumaroo 1983). In a study of human peripheral blood lymphocytes, DCM again induced chromosome aberrations in the presence and absence of a metabolic activation system when tested at 1 to 5 pL/mL (-S9) and at 2 to 6 /*L/mL (+S9). No evidence of chromosome abnormalities were seen in bone marrow cells of rats exposed by inhalation to 500, 1,500, or 3,500 ppm DCM for 6 h/day, 5 days/week for 6 months (Burek et al. 1980). When DCM was evaluated to assess its potential to induce sister chromatid exchanges (SCE), negative results were obtained following 2-, 4-, and 10-h exposures (McCarroll et al. 1983). Following a 24-h exposure to a 1.8, 3.6, 5.4, or 7% (DCM/air, v/v) atmosphere, dose-related increases (? < 0.01) of SCE were obtained at the 7% dose level (McCarroll et al. 1983). An additional study (Jongen et al. 1981) reported a weak effect on SCE induction, which reached a plateau at the 1% dose level. Exposure to concentrations as high as 4% over 1, 2, and 4 h with and without an S9 fraction did not increase the rates of SCE above those seen at 1%. ------- lexicological Data 63 Table 4.7. Results of mettaylcw chloride Bf in tartan short-tcfB tests End point Target cell Result" Reference* Chromosome aberrations Human peripheral lymphocytes CHO cells Mouse lymphoma L5178Y Rat bone marrow (microsomes) Sister chromatid exchange Human peripheral lymphocytes CHO cells Thilagar et al. 1984a Burek et al. 1984 Thilagar et al. 1984b V79 Mouse lymphoma L5178Y Point mutations L5178Y/TK CHO/HGPRT V79/HGPRT DNA damage and repair Primary rat hepatocytes Human penpheral lymphocytes Primary human fibroblastt (AH) V79 In vivo/in vitro rat hepatocytes Micronucleus In vivo/in vitro mouse hepatocytes Mouse bone marrow Thilagar et aL 1984 Jongen et aL 1981 CEFIC 1986c Perocco and Prodi 1981 CEFIC 198«e CEFIC I986d Gocke et aL 1981 CEFIC I986e a-f — positive, — — negative, ± • margmaL ------- 64 Section 4 Methylene chloride was not mutagenic In three studies utilizing several mammalian cell systems. Metabolic activation was used. Mechylene chloride did not induce unscheduled DNA synthesis (UDS) in two of three studies with primary rat hepatocytes (Andrae and Wolff 1983, CEFIC 1986c). A positive response was only marginal in the third study (Thilagar et al. 1984b). Methylene chloride was also negative in UDS studies in primary mouse hepatocytes, human peripheral lymphocytes, primary human fibroblasts, and V79 cells. When rats or mice were exposed to 2000 or 4000 ppm DCM by inhalation for either 2 or 6 h, no UDS was detected in either species (CEFIC 1986c). Methylene chloride did noc induce micronuclei in mouse bone marrow cells following two intraperitoneal administrations (at 0 and 24 h), with smears being prepared at 30 h (Gocke et al. 1981). Methylene chloride did not induce a biologically or statistically significant increase in polychromatic erythrocytes containing micronuclei when tested orally in corn oil at dose levels of 4,000, 2,500, and 1,250 mg/kg in C57BL/6J/ALpk mice (CEFIC 1986e). The induction of S-phase hepatocytes in B6C3F1 mouse liver was measured after one or two inhalation exposures to 4,000 ppm DCM for 2 h (CEFIC 1986d). The author reported small, but statistically significant, increases in the number of S-phase cells. Since the induction of S-phase hepatocytes is not a commonly used assay, the usefulness of these findings is limited. Results were equivocal in UDS studies conducted in vitro (CEFIC 1986c). The in vivo interaction of DCM and its metabolites with F344 rat and B6C3F1 mouse lung and liver DNA was measured after inhalation exposure of 4,000 ppm 14C-DCM for 3 h (CEFIC 1986b). The DNA was isolated from the tissues 6, 12, and 24 h after the start of exposure and then analyzed for total radioactivity and the distribution of radioactivity within enzymically hydrolysed DNA samples. Covalent binding to hepatic protein was also measured. An additional-group of rats and mice were dosed intravenously with ^C-formate (after exposure to nonradiolabeled DCM for 3 h) to determine the pattern of labeling resulting from the incorporation of formate into DNA via the C-l pool. Low levels of radioactivity were found in DNA from the lungs and livers of both rats and mice exposed to 14C-DCM. Two- to fourfold higher levels were found in mouse DNA and protein than in the rat. Chromatographic analysis of the DNA nucleosides showed the radioactivity to be associated with the normal constituents of DNA. No peaks of radioactivity were found that did not coincide with peaks of radioactivity present in hydrolysed DNA from formate-treated rats and mice. Under the conditions of this study, there was no evidence for alkylation of DNA by DCM, and it was concluded that DCM was not genotoxic (CEFIC 1986b). There is clear evidence of mutagenicity in bacteria. Results were mixed in tests of yeast, drosophila, and mammalian cells in cultures, and were largely negative in mammalian cells in vivo. Given the evidence of in vitro clastogenicity DNA binding studies, it was concluded that DCM may be a weak mutagen in mammalian systems (EPA 1987a,b). ------- Toxicologies! Data 65 4.3.7 Carcinogeniclty 4.3.7.1 Overview Two cohort studies examined the mortality incidence in workers occupationally exposed to DCM. Neither study reported a statistically significant increase in deaths from cancer among workers exposed to DCM These occupational studies are not adequate to rule out a risk of cancer to humans from DCM, and the compound should be regarded as though it has some potential to cause cancer. Carcinogenic responses were reported in various animal studies following inhalation and ingestion of DCM. Lung and liver tumors were reported in mice of both sexes following exposure through inhalation for lifetime. Inhalation studies involving rats and hamsters revealed sarcomas in the salivary gland region of male rats, but no malignant responses were reported in female rats and in hamsters of both sexes. The authors suggested that a salivary gland viral disease in rats may have contributed to the development of the tumorigenic response. Rats of both sexes have shown an elevated incidence of benign mammary tumors. Some liver tumors were produced in rats and mice that ingested DCM in drinking water for 2 years, but the tumor incidences were not considered by the authors to be compound related. These responses were considered by EPA to be borderline evidence of carcinogenicity. Based on the weight of evidence from these animal studies, and applying the criteria described in EPA's guidelines for assessing carcinogenic risk (EPA 1986c), DCM was classified in Group B2: probable human carcinogen (EPA 1985b). 4.3.7.2 Human Data were found in the available literature for inhalation exposure, but not for the oral or dermal administration of DCM. Friedlander et al. (1978) analyzed mortality statistics for male workers exposed to low levels of DCM. Measurements from 1959 to 1976 were TWA concentrations in the range of 40 to 120 ppm (estimated), both from air monitoring and blood CO-Hb levels for up to 30 years. No excess cancer mortality was found compared with the unexposed population. Hearne et al. (1987) evaluated an expanded employee cohort for cancer mortality. A maturing 1964 to 1970 cohort of 1,013 hourly men were evaluated through 1984. On average, employees were exposed at a rate of 26 ppm for 22 years (median latency was 30 years). Compared with the general population, no statistically significant excesses were observed in deaths from malignant neoplasms (41 observed vs 52.7 expected), from respiratory cancer deaths (14 vs 16.6), or from liver cancer deaths (0 vs 0.5). An increased incidence of pancreatic cancer deaths (8 vs 3.1 expected) was reported; however, these deaths were not considered to be statistically significant (Hearne et al. 1987). Ott et al. (1983a) evaluated cancer mortality among employees of a fiber production plant who were exposed to TWA concentrations of 140 to 475 ppm. They observed fewer than expected deaths from malignant neoplasms in comparison to expected deaths from the same cause in the general population. ------- 66 Section 4 4.3.7.3 Animal Several studies were found in the available literature that demonstrated the carcinogenic potential of DCM (Table 4.8). Risk estimates were conducted by EPA based on the occurrence of liver and lung tumors following the inhalation of DCM by mice (NTP 1986) and on suggestive evidence in mice (NCA 1983) following the ingestion of DCM-contaminated drinking water. The NTP (1986) reported that in mice exposed co DCM, the incidence of hepatocellular carcinomas alone was statistically increased over controls in high-dose males (P - 0.005) and in both high-dose females (P < 0.001) and low-dose females exposed to 0 (male 13/50, female 1/50), 2000 (male 15/49, female 11/48), and 4,000 ppm (male 26/49, female 32/48) for 102 weeks. The incidence of combined hepatocellular adenomas and carcinomas in the 4,000-ppm group of male mice (33/49) was significantly (P - 0.02) higher than controls (22/50). There were also increased incidences in the low-dose (16/48, P < 0.001) and high-dose (40/48, P < 0.001) females when compared with controls (3/50). Methylene chloride induced a dose-dependent statistically significant increase in lung adenoma and carcinoma in mice of both sexes exposed through inhalation for 2 years (NTP 1986). Tumor incidences were as follows: at 2,000 ppm, 30 of 48 female mice and 27 of 50 male mice developed lung tumors compared with 3 of 50 (female) and 4 of 50 (male) in the control groups. At 4,000 ppm, 41 of 48 female mice and 40 of 50 male mice developed lung tumors. The NCA (1983) evaluated the carcinogenicity of ingested DCM in B6C3F1 mice from drinking water at doses of 0, 60, 125, 185, or 250 ing/kg/day £°r 104 weeks. The incidence of proliferative hepatocellular lesions was higher in treated males than in male control groups (but not in females); however, the increase was reported not to be dose related or statistically significant when ompared with concurrent control rates. In addition, the incidence of lesions in treated groups was within the historical range of control values. It was concluded, therefore, that DCM did not induce a carcinogenic response under the study conditions. An independent assessment of the NCA (1983) data by the EPA (1985a) concluded that the incidence of heptocellular adenomas and carcinomas (combined) was significantly (P < 0.05) increased in male B6C3F1 mice exposed to 125 mg/kg/day (30/100) or 185 mg/kg/day (31/99) of DCM in drinking water when compared with controls (24/125). The EPA (1985a) concluded that, on the basis of the increased incidence observed in the NCA study, DCM administered in deionized drinking water at doses up to 250 mg/kg/day produced borderline carcinogenicity in B6C3F1 male mice. In a rat study (NCA 1982), DCM was administered in drinking water to both sexes at 0, 5, 50, 125, and 250 mg/kg/day for 104 weeks. An increase in liver tumors was reported in females treated at 50 and 250 ngAg/day when compared with concurrent controls. These levels were within historical control ranges. The authors concluded that DCM was not carcinogenic under conditions of the study. The EPA (1985a) evaluated results of the study and reported an increased incidence of neoplastic nodules combined with hepatocellular carcinomas in female rats ------- Toxicologies! Data 67 Table 4.8. Summary of primary tumors in rats and mice in 2-year studies of methyleoe chloride Route Species Strain Concentration Tumor site References Inhalation Inhalation Drinking water Drinking water Inhalation Inhalation Hamster Rat Rat Mouse Rat Mouse Syrian Sprague-Dawley F344 B6C3F, F344/N B6C3F, 0, 500, 1,500, 3,500 ppm 0, 50, 200, 500 ppm 0, 5, 50, 125, 250 mg/kg 0, 60, 125, 185, 250 mg/kg 0, 1,000, 2,000, 4,000 ppm 0, 2.000, 4,000 ppm No reported effect Mammary gland (females) Liver (female) Liver (male) Mammary gland (both sexes) Liver and lung (both sexes) Nitschke et al." 1982 National Coffee Association 1982 National Coffee Association 1983 National Toxicology Program 1986 •Nitschke efal. 1988. Source: Adapted from NTP 1986. ------- 68 Section 4 (P < 0.05) when compared with matched controls. Tumor incidences were as follows 0/134, 1/85, 4/85, 1/85, and 6/85 in combined control, 5-, 50-, 125-, and 250-mg/kg/day groups, respectively. The EPA (1985a) concluded that based on the combined incidence of adenomas and carcinomas, DCM showed borderline evidence of carcinogenicity in rats when administered orally. Burek et al. (1980, 1984) exposed rats and hamsters of both sexes to DCM (0, 500. 1,500, or 3,500 ppm) via inhalation for 2 years. Sarcomas in the salivary gland region of male rats were reported at 1,500 or 3.500 ppm. This effect was statistically significant in the 3,500-ppm group, with a trend (numerically increased but not statistically significant) for an increase in the 1,500-ppm group. The toxicological significance of this finding is unknown. Results were negative for female rats and for hamsters of both sexes. The authors suggested that a salivary gland viral disease in rats may have contributed to the tumorigenic response. Rats of both sexes were administered DCM (at doses of 0, 50, 200, or 500 ppm) via inhalation for 2 years (Nitschke et al. 1982, 1988). Female rats exposed to 500 ppm had an increased (P < 0.05) number of spontaneous benign mammary tumors (tumor-bearing rat adenomas, fibromas, and fibro adenomas, with no progression toward malignancy). The incidence of benign mammary tumors in female rats exposed to 50 or 200 ppm was comparable to historical control values. No increase in the number of any malignant tumor types was observed in rats exposed to concentrations as high as 500 ppm DCM. 4.4 INTERACTIONS WITH OTHER CHEMICALS Limited studies were found in the available literature on the interactions of DCM with other chemicals. Much of the data found focused on concurrent exposure to carbon monoxide, since DCM is metabolized to carbon monoxide. In rats, combined carbon monoxide and DCM exposure yielded additive increases in the carboxyhemoglobin levels (ACGIH 1986) Fodor and Roscavanu (1976) reported that exposure of human subjects to 500 ppm of DCM resulted in levels of carboxyhemoglobin in the blood comparable with those produced by the Threshold Limit Value for carbon monoxide (50 ppm). Because carbon monoxide generated from DCM is additive to exogenous environmental carbon monoxide, DCM exposures at high levels could pose an additional human health burden. Of particular concern are workers, smokers (who maintain significant constant levels of carboxyhemoglobin), and others who may have increased sensitivity to carbon monoxide toxicity, including persons with cardiovascular disease and respiratory dysfunction. ------- 69 5. MANUFACTURE. IMPORT, USE, AND DISPOSAL 5.1 OVERVIEW Approximately 265,000 megagrams (Mg) of DCM were produced in the United States in 1983 at six major production plants in four states. After accounting for imports and exports, about 252,000 Mg were available for U.S. consumptive use (50 FR 42037). The major uses of DCM are in aerosol products and as a paint remover, which together account for more than 50% of the DCM used in the United States in 1983. Although data on land disposal of DCM from production processes were not available, it appears that land disposal is not a major source of the DCM released to the environment. 5.2 PRODUCTION Methylene chloride is produced in the United States by two major processes. The methane chlorination process combines methane and chlorine in a closed reactor. The DCM and carbon tetrachloride are separated from the reaction mixture by distillation. The second process, chlorination of methyl chloride, combines methyl chloride (formed by reaction of hydrogen chloride with methanol) and chlorine in a reactor vessel. The process flow is passed through a hydrochloric acid stripper Both these processes allow recycling of hydrochloric acid (OSHA 1986, HSDB 1987). There were four major producers of DCM in the United States in 1983, with plants at six locations in four states (Texas, West Virginia, Louisiana, -and Kansas). The total 1983 production volume was approximately 265,000 Mg (OSHA 1986, EPA 1985d). 5.3 IMPORT In 1983, 20,000 Mg of DCM were imported into the United States, and 33,000 Mg were exported (50 FR 42038, OSHA 1986). 5.4 US» The two major consumptive uses of DCM in the United States in 1983 were as a paint remover and as a solvent and flammability depressant in aerosol products such as coatings, paint removers, hair sprays, room deodorants, herbicides, and insecticides. Of the total U.S. production of DCM in 1983 (265,000 Mg), 69,400 Mg (26.2%) were used in aerosol products, and 63,100 Mg (23.8%) were used in paint removal products. Other important uses of DCM include foaa blowing of polyurethanes, mecal degreasing, stripping and degreasing in the electronics industry, as a solvent in polycarbonate resin production, in photographic film-base ------- 70 Section 5 manufacturing, adhesive production, and numerous other solvent. cleaning, and thinning uses. Methylene chloride is also occasionally used as an extractant for caffeine, spices, and hops (OSHA 1986, HSDB 1987, EPA 1985a). 5.5 DISPOSAL No data were found on the land disposal of DCM from production processes. However, it appears that land disposal is not a major route of the environmental release of DCM (EPA 1985a). ------- 71 6. ENVIRONMENTAL FATE 6.1 OVERVIEW Methylene chloride is released Co Che environmenc mainly through its varied and widespread uses. PainC removal, aerosol use, mecal degreasing, foam blowing, and miscellaneous uses account for"the bulk of DCM releases. Approximately 85% of the total amount of DCM produced in the United States is released to all media, primarily in industrialized areas. Although no estimates of releases to specific media are available, most is probably released to the air. The chlorination of drinking water also produces DCM. Volatilization of DCM occurs from all sources and in all environmental media. While atmospheric transport and dispersion are important, degradation by hydroxyl radicals, photolysis, and intermedia transfer via rainout prevent DCM from accumulating in the atmosphere. Because of its moderate water solubility, leaching from soil and transport in groundwater and surface water are important fate processes. Soil adsorption and abiotic transformation processes in aquatic systems are not major factors for DCM. Biodegradation may be an important fate process, but bioaccumulation and bioconcentration are not considered to be significant factors in aquatic, sediment, and wastewater treatment systems. 6.2 RELEASES TO THE ENVIRONMENT 6.2.1 Anthropogenic Sources The major sources of DCM are anthropogenic, as are DCM emissions to the environment. Although many varied and widespread uses contribute to DCM emissions, paint removal, aerosol use, metal degreasing, foam blowing, and miscellaneous uses account for 158,900 Mg/year (about 81%) of the -196,000 Mg/year released (50 FR 42037, Oct. 17. 1985). These releases occur primarily to sewage treatment plants, surface water, land, and the atmosphere (EPA 1985a). Losses during production, transport, and storage are primarily fugitive (e.g., from leaky Joints, values, and pump seals). These losses are poorly documented; however, they are considered to be a small percentage of the total emissions from other uses (EPA 1985a). The maximum concentration measured in industrial wastewater (210 ppm) is associated with discharges by the paint- and ink- formulating and aluminum-forming industries (EPA 1981a, as cited in HSDB 1987). Other industries in which concentrations of DCM exceed an average discharge concentration of 1 ppm include coal mining, photographic equipment and supplies, pharmaceutical manufacturing, organic chemical and plastics manufacturing, rubber processing, and laundries. Methylene ------- 72 Section 6 chloride is produced in drinking water (in Che low parts-per-billion concentration range) during chlorination treatment (NAS 1977 Bellar et al. 1974, as cited in EPA 1985a). 6.2.2 Natural Sources Methylene chloride may be formed from natural sources, but these are not believed to contribute significantly to global releases (NAS 1978, as cited in EPA 1985d). 6.3 ENVIRONMENTAL FATE 6.3.1 Atmospheric Fate Processes Methylene chloride released to the atmosphere is expected to readily disperse and be transported some distance from its source. It is not expected to accumulate significantly in the atmosphere, however. because of the reaction with hydroxyl radicals (OH-). This reaction is considered the primary tropospherlc chemical scavenging process for DCM (EPA 1985a). Based on reaction rate studies summarized by the EPA (1985a), the lifetime of DCM in the troposphere ranges from a minimum of a few months to a maximum of 1.4 years under typical U.S. conditions (Altshuller 1980; Cox et al. 1976; Crutzen and Fishman 1977; Davis et al. 1976; Singh 1977; Singh et al. 1979, 1983). Most estimates of how long DCM lingers in the atmosphere are less than a year, suggesting atmospheric accumulation is not important, whereas dispersive transport is a matter of concern. Butler et al. (1978, as cited in EPA 1985a) proposed that the reaction of DCM and OH- may form phosgene (COC12) as a degradation product. The small amount of DCM that reaches the stratosphere (about 1%) will degrade via photolysis and reaction with chlorine radicals (HSDB 1987). The highest concentrations of DCM have been observed at night and in early morning (Singh et al. 1982, as cited in HSDB 1987), which suggests that photooxidation may be an important degradation process. Pearson and McConnell (1975) reported on laboratory studies that show that the photolysis of DCM may yield O>2 and hydrochloric acid. 6.3.2 Surface Vater/Groundvater Fate Processes Volatilization is the most important fate process In surface waters, whereas transport is likely to be important in groundwater, considering DCM's moderate water solubility. Dewalle and Chian (1978) and Helz and Hsu (1978) (as cited in HSDB 1987) reported DCM as nondetectable 3 to 15 miles downstream from where it had been released into a river. The rate of volatilization from surface water is expected to be strongly affected by wind and mixing conditions. Biodegradation may be an important fate process under both aerobic and anaerobic conditions. Hydrolysis occurs slowly in this medium and LS strongly influenced by pH and temperature. Studies cited by EPA (1985c) indicate an estimated half-life of 18 months at 25°C. ------- Environmental Face 73 6.3.3 Soil Fate Processes Volatilization, leaching, and biodegradation are expected to be important fate processes for DCH in soil. Adsorption to soil is not expected to be important (Dilling et al. 1975, as cited in EPA 198Sc). 6.3.4 Biotic Fate Processes The EPA (198Sa) cites a number of studies indicating the microbial (e.g., Pseudomonas spp.) biodegradation of OCM in aquatic systems (Brunner et al. 1980, Lapat-Polasko et al. 1984, Rittman and HcCarty 1980, Klecka 1982, Wood et al. 1981). Breakdown appears to yield methyl chloride as an intermediate and C02 as the final carbon-containing end product. Modeling indicates the biodegradation rate is about 12 times greater (in a continuously stirred activated sludge reactor containing acclimated microorganisms) than the volatilization rate (Klecka 1982, as cited in EPA 198Sa). HSDB (1987) reports biodegradation rates for sewage seed or activated sludge ranging from 6 h to 7 days under aerobic conditions, based on results of five studies (Davis et al. 1981, Klecka 1982, Rittman and HcCarty 1980, Stover and Kincannon 1983, Tabak et al. 1981). Hansch and Leo (1979, as cited in HSDB 1987) report a low octanol/vater partition coefficient for DCM (log P - 1.25), suggesting bioaccumulation and bioconcentration are not important fate processes. ------- 75 7. POTENTIAL FOR HUMAN EXPOSURE 7.1 OVERVIEW Host human exposure to DCM occurs via air. Air exposures are highest in occupational settings and near sources of emissions. Inhalation exposures are also higher in industrialized urban areas. There is a potential for low-level general population exposure from ambient DCM emissions from paint removal, aerosol use, metal degreasing, electronics, Pharmaceuticals, food processing, and miscellaneous uses. Hazardous waste sites release DCM to the air, groundwater, and surface water. Very low levels of DCM exposure occur through ingestion of decaffeinated coffee. Monitoring data on DCM exposure levels outside of occupational settings are sparse. Populations at elevated risk for*exposure to high concentrations of DCM are individuals in occupational settings in the following industrial categories: DCM manufacturing, paint remover formulation, polycarbonate resin production, and cleaning solvent. Consumers may be exposed to significant amounts of DCM vapors through the use of a variety of products. These are summarized in Sect. 7.4. 7.2 LEVELS MONITORED OR ESTIMATED IN THE ENVIRONMENT 7.2.1 Levels in Air Methylene chloride occurs in ambient air throughout the United States, with background levels reported in a number of studies falling in the range of 30 to 50 ppt (Brodzinsky and Singh 1983, Cronn et al. 1977, Cronn and Robinson 1979, Grimsrud and Rasmussen 1975, Rasmussen et al. 1979, Robinson 1978, Singh et al. 1983, as cited in EPA 1985a). Measured ambient air levels (as mixing ratios) of DGM (Grimsrud and Rasmussen 1975; Pellizzari 1977, 1978a, 1978b; Pellizzari and Bunch 1979; Pellizaari et al. 1979; Rasmussen et al. 1979; Singh et al. 1979, 1980, 1981) are summarized by the EPA (1985a). The data confirm maximum and mean concentrations in urban areas are the highest, with Front Royal, Virginia, and Edison, New Jersey, having the highest values for urban areas. Pellizzari and Bunch (1979, as cited in EPA 1985a) reported the highest ambient air level of DCM in a New Jersey survey to be 360 ppb. Measurements were made during an 11-min sampling period at a waste disposal site in Edison. Harkov et al. (1985) reported average air concentrations of DCM at six abandoned hazardous waste sites and at one sanitary landfill in New Jersey to fall in the range of 0.1 to 12.3 ppb, with a maximum measured concentration of 53.8 ppb. ------- 76 Section 7 7.2.2 Levels in Water Methylene chloride occurs in drinking water, surface water, and groundwater throughout the United States. Six of seven studies reviewed by EPA (1985d) reported the presence of DCM in drinking water (Bellar et al. 1974; Coleman et al. 1976; Dowty et al. 1975; Pellizzari and Bunch 1979; EPA 1975a, 1975b). Mean reported concentrations in finished drinking water were around 1 pg/L or less, with maximum concentrations reported at <3 A»g/L. Bellar et al. (1974, as cited in EPA 1985a) reported 2.0 ng/L DCM in finished water produced from raw river water containing no detectable DCM. Dyksen and Hess (1982, as cited in HSDB 1987) reported that 2% of the drinking water samples in a ten-state survey of groundwater supplies were positive for DCM, with a maximum reported concentration of 3.6 ppm. Methylene chloride also occurs in commercially bottled artesian well water (Dowty et al. 1975, as cited in EPA 1985a). Ewing et al. (1977, as cited in EPA 1985a) reported DCM at concentrations greater than 1 ppb in 32 of 204 surface water sites sampled in heavily industrialized river basins. Pellizzari and Bunch (1979, as cited in EPA 1985a) reported a mean DCM concentration of 2.6 Pg/L and a maximum of 15.8 pg/L in untreated Mississippi River water in Jefferson Parrish, Louisiana. The EPA (1981b, as cited in EPA 1985a) reported DCM levels in EPA's automated database (STORET) files for the period 1978 to 1981 to range from 0 to 120 /ig/L in general ambient waters. Sediment concentrations were in the range of 427 to 433 ppb in 60 of 118 cases. Values in surface waters from the Ohio River Valley and Great Lakes region fall in the range of 1 to 30 ppb (ORVWSC 1982, Konasewich et al. 1978, as cited in HSDB 1987). Most of the samples with detected concentrations of DCM from the Ohio River Valley (219 of 238) were below 10 ppb. Methylene chloride has been detected in groundwater at levels up to 8.4 mg/L at 36 Superfund hazardous waste sites and in surface water at 17 other waste sites. Methylene chloride is ranked 16th of the 25 substances most frequently detected at the 53 Superfund sites sampled by EPA (1985e and 1985f, as cited in 50 FR 42037, Oct. 17, 1985). 7.2.3 Levels in Soil The EPA (1981b, as cited in EPA 1985a) reports that measurements of ambient DCM soil concentrations (for areas not associated with hazardous waste sites) are not available. 7.2.4 Levels in Food Information on levels of DCM in food is generally not available. The Food and Drug Administration has placed a limit of 10 ppm DCM for decaffeinated coffee (50 FR 51551, Dec. 18. 1985). Measured amounts are significantly less than this limit. 7.2.5 Resulting Exposure Levels The highly variable atmospheric concentrations of DCM in different areas make any quantitative estimates of exposure levels highly ------- Potential for Human Exposure 77 speculative and somewhat meaningless. Exposures from drinking water (being more relevant to hazardous waste site environmental contamination situations) can be roughly estimated from data summarized earlier. The mean concentration of DCM in finished drinking waters (from all sources) was reported at about 1 pg/L with maximum concentrations <3 /ig/L. This would result in a daily exposure of about 2 /jg/day (mean) and <6 jig/day (maximum) for a 70-kg adult with an assumed water consumption of 2 L/day. Since concentrations up to 8.4 mg/L DCM have been detected in groundwater at hazardous waste sites, an upper range estimate of localized human exposure is 16.8 mg/day. This estimate assumes that the sole source of drinking water is the contaminated aquifer, and it is used directly without treatment for the removal of DCM. The FDA's limit of 10 ppm DCM in decaffeinated coffee is estimated to result in a lifetime average exposure of 140 fig/day for consumers of brewed (roasted and ground) decaffeinated coffee, and 55 pg/day for consumers of instant decaffeinated coffee (50 FR 51551, Dec. 18, 1985) Insufficient data are available to estimate exposure from the intake of other foods. Occupational inhalation exposure levels are summarized in Sect. 7.3. Data on other exposure levels are not available. 7.3 OCCUPATIONAL EXPOSURES Occupations in which exposure to DCM may occur are listed in Table 7.1. Nearly 2.5 million workers are estimated to be exposed to DCM (ICAIR 1985). Highest occupational inhalation exposure levels (1,750 mg/nr*, as an 8-h TWA) occur in the following industrial categories (Table 7.2): DCM manufacturing (production workers), paint remover formulation (production filing), and polycarbonate resin production (production worker) (50 FR 42037, Oct. 17, 1985). The industry-wide 8-h TWA exposure level is 22.4 ppm (51 FR 42257, Nov. 24,1986). High exposure concentrations may develop rapidly in poorly ventilated areas. The EPA (50 FR 42037, Oct. 17, 1985) reports breathing zone exposures of 1,000 to 3,000 ppm DCM in simulated consumer exposure to paint strippers in a room-sized environmental chamber. 7.4 CONSUMER EXPOSURE The Consumer Product Safety Commission (CPSC) (1987) reported that products in the following classes (that contain DCM) can expose consumers to significant amounts of DCM vapors and are thus hazardous: paint strippers, adhesives and glues, paint thinners, glass frosting and artificial snow, water repellants, wood stain and varnishes, spray paint, cleaning fluids and degreasers, aerosol spray paint for automobiles, automobile spray primers, and products sold as DCM. The commission stated that it does not have up-to-date information showing that products in these categories that contain 1% or less of DCM are hazardous substances. ------- 78 SeccIon 7 Table 7.1. Occupations in which methylene chloride exposures may occur Aerosol packagers Bitumin makers Fat extractors Leather finish workers Paint remover makers Solvent workers Production workers in methylene chloride manufacturing Mold release workers Paint strippers Foam operators in the polyurethane foam blowing industry Extrusion workers in the triacetate extrusion business Assemblers and other workers in the plastics industry Printers and other workers in the printing industry Maintenance workers Painters (including spray painters) Machine operators and assemblers using cleaning solvents Anesthetic makers Degreasers Flavoring makers Oil processors Resin makers Stain removers Packers in aerosol packing Production filling and paint remover formulators Solvent reclaimers Production workers with pharmaceutical solvents Production workers in film-base manufacturing Laminators Gluers and other workers using adhesives Plate-makers Compounders of production solvents Assemblers and other workers in the electronics industry Production workers in the polycarbonate resin production industry Source: ICAIR 1985. ------- Potential for Human Exposure 79 Table 7.2. Summary of occopadoaal exposure to netayteae chloride Industry category Methylene chloride manufacturing Aerosol packing Aerosol use Paint remover formulation Paint remover use Polyurethane foam blowing Metal degreating Solvent recovery Pharmaceutical solvent Triacetate extrusion Solvent for plastics Adhesive use Printing Production solvent Electronic Cleaning solvent Polycarbonate rain production Not classified Job category Production worker Packer Spray painter Mold release Other workers Production Tilling Paint stnpper Water wash Other workers Foam operator Other worker Degreaser Solvent reclaimer Production worker Extrusion Finish Other worker Gluer Assembler Other worker Laminator Gluer Other worker Printer Plate- maker Other worker Compounder Other worker Assembler Other worker* Maintenance Painter Machine operator Assembler Production worker Other worker Estimated number of workers 1.200* 896r 14.296 25.972 50,343 1.280 7.680* 25.60T/ 3.840* 405e 162' 227.848 170* 19.046 340 460 120 1.514 984 8.022 1.366 280 901 43,931 15.911 27.531 15,041 1.531 10.399 11.267 49.884 1.831 79.951 22.252 1.200 342,064 1.016,894 8-h TWA exposure (mg/m3)" 1.750 317 46 80 28 1.750 57 67 192 51 97 46 13 141 1.479 187 1.036 437 197 21 30 6 15 17 32 56 44 8 14 29 30 32 30 1,000 1.750 59 Inhalation exposure (mg/day)* 17.500 3.172 462 798 "277 17,500 572 673 1.930 513 974 464 128 1.408 14,795 1.869 10.363 4,368 1.966 211 295 57 153 173 324 561 439 82 135 292 304 317 297 9,999 17,500 592 "Values are based on the geometric mean of the monitoring samples reported for a given job Values are based on the assumption that the breathing rate for workers is 10 m /day "Estimated number of workers based on average number of workers per plant multiplied by the total number of plants identified. The 'not classified' category represents data obtained from many types of industrial plants which are not well defined. Source: Adapted from 50FR42037. October 17. 1985. ------- 80 Section 7 7.5 POPULATIONS AT HIGH RISK 7.5.1 Above-Average Exposure From the previous discussion, it is evident that a diverse number of occupational subpopulations representing more than one million workers are at risk for receiving DCM exposures substantially above the general population. Among the general population, transitory high-level atmospheric. and sometimes dermal, exposures to DCM are commonly associated with the use of DCM-containing paint strippers, adhesives, and aerosol products for hobby and household uses. Further, the general widespread. distribution of DCM throughout the atmosphere contributes additional exposures to the general population that are usually classified as low level, but may become significant in the vicinity of DCM industrial and point-of-use areas (especially in urban areas). Populations downstream from DCM production or use facilities may be at increased risk of exposure from discharges into surface waters. Also, populations near hazardous waste sites should be considered at high exposure risk, especially if their drinking water supply Is derived from local groundwater. Certain products that contain DCM can expose consumers to significant amounts of vapors and are thus hazardous. These have been summarized in Sect. 7.4. The CPSC (1987) stated that it does not currently have data showing that products in these categories that contain 1% or less of DCM are hazardous substances. 7.5.2 Above-Average Sensitivity Persons with compromised cardiovascular systems are considered to be more sensitive to DCM exposure. ------- 81 8. ANALYTICAL METHODS Gas chromatography (GC) is Che mosc common analytical method for detecting and measuring DCH in environmental and biological samples. Samples may be prepared by several methods, depending on the matrix being sampled. The preparation procedures for analyzing environmental samples for DCM include charcoal adsorption/desorption, purge and trap, headspace sampling, and vacuum distillation (EPA 1983, 1986b; APHA 1977, Page and Charbonneau 1977, 1984). Preparation of biological samples often involves headspace sampling (DiVincenzo et al. 1971). The GC separates complex mixtures of organics and allows individual compounds to be identified and quantified. Detectors used to identify DCH include a flame ionization detector (FID), electron capture detector (ECD), and an electrolytic conductivity detector and halogen-specific detector (HSD) (EPA 1983, 198Sa. 1986b; APHA 1977; Page and Charbonneau 1984). When unequivocal identification is required, a mass spectrometer (MS) coupled to a GC column (GC-MS) may be used (EPA 1983, 1985d; Pellizzari and Bunch 1979). Methylene chloride is a common laboratory contaminant and is frequently found in laboratory blanks. 8.1 ENVIRONMENTAL MEDIA Representative methods appropriate for measuring DCM in environmental media are listed in Table 8.1. 8.1.1 Air The American Public Health Association (APHA) method for measuring of organic solvent vapors in air (834) is accepted by NIOSH (Classification B) for application to DCM in workplace air. The method involves adsorption of the organic vapors in the air onto an activated charcoal filter, desorption with carbon disulfide, and injection of an aliquot of the desorbed sample into a GC equipped with an FID. The resulting peaks are measured and compared with standards (APHA 1977). Interferences may be caused by high temperatures, high humidity, high sampling flow rates, and other solvents with the same retention times. Different GC column materials and temperatures may be used to resolve the interference of other solvents (APHA 1977). Other methods for measuring DCM in ambient air include GC/MS and GC/ECD (EPA 1985d, Pellizzari 1974, Pellizzari and Bunch 1979). ------- 82 Section 8 Sample matrix Table 8.1. Analytical methods for methylene chloride in environmental samples Sample preparation Analytical method" Sample detection limit Accuracy* References Air Charcoal adsorption, GC/FIO desorption with carbon disulfide Air Charcoal adsorption, GC/FID desorption with carbon disulfide 25 ppbc 90-110%' APHA 1977 10 mg/m3 95.3% NIOSH 1984 Water Soil Food Purge and trap Purge and trap Purge and trap Purge and trap or direct injection Headspace sampling Vacuum distillation Vacuum distillation GC/HSD EPA method 601 GC/HSD EPA method 624 GC/MS EPA method 8010 GC/HSD GC/ECD GC/ECD GC/EC 0.5 Mg/L« 0.25 Mg/L 2-8 Mg/L ND^ 0.05 ppm 7ng 7ng 104% 97.9% 96% ND ND 94% 100% APHA 1985 EPA 1983 EPA 1983 EPA 1986b Page and Charbonneau 1984 Page and Char-onneau 1977 Page and Charbonneau 1977 "GC - gas chromatography; FID - name ionization detector, HSD = halogen-specific detector: MS — mass spectrometry, BCD — electrolytic conductivity detector. EC - electron capture detector * Average percent recovery. 'Lowest value for various detected compounds reported during collaborative testing. 'Estimated accuracy of the sampling and analytical method when the personal sampling pump is cali- brated with a charcoal tube in the line. 'For most halogenated methanes and ethanes; not specific for DCM. /Not determined. ------- Analytical Methods 83 8.1.2 Water The EPA approved two methods (601, 624) for analyzing wastevater samples for DCM. These methods use a packed-column GC for separating organic pollutants, but they differ in sample preparation procedures and detection instrumentation. Method 601 is optimized for purgeable halocarbons with a detection limit of 0.25 A«g/L for DCM. Method 624, which employs MS for detection and quantification, is broadly applicable to a large number of purgeable organics and has a detection limit of 2.8 pg/L for DCM. The APHA standard method for halogenated methanes and ethanes (514), like EPA Method 601, uses purge-and-trap sample preparation and a halogen- specific detector. 8.1.3 Soil Soil samples can be analyzed for DCM by GC with a halogen-specific detector. Sample preparation is by purge and trap. This method (8010) is approved by the EPA for analysis of halogenated volatile organics in solid waste. The method detection limit for DCM has not been determined (EPA 1986b). 8.1.4 Food Methylene chloride in food products (e.g., decaffeinated coffee, spice, and oleoresins) may be measured by GC/ECD or GC with an electrolytic conductivity detector (Page and Charbonneau 1984, 1977; Page and Kennedy 1975). A GC/MS method may also be used for food analysis (Hiatt 1981). 8.2 BIOMEDICAL SAMPLES Methods appropriate for measuring DCM in biological samples are listed in Table 8.2. 8.2.1 Fluids and Ezudates Blood levels of DCM may be measured using GC with an FID. Headspace sampling eliminates the need for an extraction procedure (DiVincenzo et al. 1971). Infrared analysis has also been used for quantitative estimation of volatile organics in blood, but the infrared method requires an extraction step (DiVincenzo et al. 1971). Analysis of urine for DCM is by the sane method used for blood. Breath samples were also analyzed for DCM by GC/FID (DiVincenzo et al 1971). 8.2.2 Tissues Human adipose tissue levels of DCM may be measured by GC/FID, using hydrochloric acid to break down the adipose tissue and then a headspace sampling method (Engstrom and Bjurstrom 1977). ------- 84 Section 8 Table 8.2. Analytical methods for methylene chloride in biological samples Sample Sample Sample Analytical detection matrix preparation method" limit Accuracy References Blood Headspace GC/FID 0.022 mg/L 49.8% DiVincenzo et al. 1971 sampling Urine Headspace GC/FID ND* 59% DiVincenzo et al. 1971 sampling Breath Gas sample GC/FID 0.2 ppm ND DiVincenzo et al. 1971 valve Adipose Acid GC/FID 1.6 mg/kg* ND Engstrom and Bjurstrom tissue hydrolysis 1977 headspace sampling "GC = gas chromatography; FID = flame iomzation detector. *ND = not determined. c Lowest reported concentration. ------- 85 9. REGULATORY AND ADVISORY STATUS 9.1 INTERNATIONAL The World Health Organization (WHO) has not recommended a drinking water guideline value for DCM. 9.2 NATIONAL 9.2.1 Regulations Regulations applicable to DCM address occupational exposure concentrations; reporting requirements; spill quantities; presence In food, cosmetics, and hazardous waste; and tolerance requirements exemptions (Table 9.1). The Occupational Safety and Health Administration (OSHA) sets Permissible Exposure Limits (PELs) for occupational exposures to chemicals based on the recommendations of the NIOSH. The OSHA PELs for DCM are 500 ppm (1,737 mg/m3) in workplace air for a time-weighted average (TWA) (8 h/day, 40 h/week), a ceiling concentration of 1.000 ppm (3,474 mg/m3), and a maximum peak, not to be exceeded for more than 5 min in 2 h, of 2,000 ppm (6,948 mg/m3) (OSHA 1986). The EPA Office of Drinking Water (ODW) has promulgated monitoring regulations for 51 unregulated volatile organic compounds (VOCs), including DCM (52 FR 25690). These regulations require the monitoring of public water supplies in a program to be phased in over 4 years. The Comprehensive Environmental Response, Compensation, and Liability Act of 1980 (CERCLA) requires that persons in charge of facilities from which a hazardous substance has been released in quantities equal to or greater than its reportable quantity (RQ) immediately notify the National Response Center of the release. The RQ for DCM set by the EPA Office of Emergency and Remedial Response (OERR) is 1,000 Ib. Chemicals are included on the Resource Conservation and Recovery Act (RCRA) Appendix VIII list of hazardous constituents (40 CFR Part 261) if they have toxic, carcinogenic, mutagenic, or teratogenic effects on humans or other life forms. Methylene chloride is included on this list, and wastes containing DCM are subject to the RCRA regulations promulgated by the EPA Office of Solid Waste (OSW). The Office of Toxic Substances (OTS) promulgates regulations related to manufacturers and/or processors of chemicals that may present an unreasonable risk to health or che environment. Manufacturers of DCM are required to submit to EPA information on the quantity of the ------- 86 Section 9 Table 9.1. Regulations and guidelines applicable to methylene chloride Agency Description Value References National Regulations OSHA Permissible exposure limit (PEL) in workplace air Time-weighted average (TWA) (8 h/day, 40 h/week) Ceiling concentration Maximum peak (not to exceed 5 min in any 2 h) EPA OERR Reportable quantity (RQ) EPA OSW Hazardous constituent list Appendix VIII EPA OTS Preliminary assessment information rule Health and safety data reporting rule Comprehensive assessment information rule (proposed) FDA Ban on use of methylene chloride in cosmetics (proposed) Limit in decaffeinated roasted coffee and soluble coffee extract (instant coffee) EPA OPP Exemption from tolerance SOOppm 1,000 ppm 2,000 ppm EPA ODW Monitoring regulations for unregulated VOCs NAa 1.000 Ib NA NA NA NA NA 10 ppm NA 29CFR 1910.1000 (1971) 40 CFR 141 40 52 FR 25690 (07/08/87) 40 CFR 302.4 40 CFR 1173 SOFR 13456 (04/04/85) 40 CFR 261 45 FR 33084 (05/19/80) 40 CFR 712 47 FR 26992 (06/22/82) 40 CFR 716 47 FR 38780 (09/02/82) 51 FR 35762 (10/07/86) SOFR 51551 (12/18/85) 21 CFR 173.255 32 FR 12605 (08/31/67) 40 CFR 180.1010 36 FR 22540 (11/25/71) ------- Regulacory and Advisory Status 87 Table 9.1 (condoned) Agency NIOSH EPA ODW NAS IARC EPA ACGIH Description National Guidelines Recommended exposure limit (REL) Immediately dangerous to life or health (IDLH) Health advisories (HAs) One-day (child) Ten-day (child) DWEL* Suggested no adverse response level (SNARL) One-day Seven-day Cancer ranking Cancer ranking Threshold limit value (TLV-TWA) Value Lowest feasible limit 5,000 ppm 13.3mg/L 1.5 mg/L 1.75 mg/L 45.5 mg/L 6.5 mg/L Group 3 Group B2 50 ppm (175mg/m3) References NIOSH 1986 NIOSH 1985 EPA 1985g EPA 1985g NAS 1980 IARC 1986 EPA 1985g ACGIH 1986 "NA = not available. * DWEL =* drinking water equivalent level. ------- 88 Section 9 chemical manufactured or imported, the amount directed to certain uses, and the potential exposure and environmental release of the chemical under the Preliminary Assessment Information Rule. Unpublished health and safety studies on DCH must be submitted to EPA by manufacturers or processors under the Health and Safety Data Reporting Rule. The OTS has proposed a Comprehensive Assessment Information Rule (CAIR) that would specify the reporting requirements for chemicals for which EPA requires information. This rule is designed to streamline the collection of information to support chemical risk assessment/management strategies. Methylene chloride is one of the 47 chemicals proposed to be included in this rule. The OTS has initiated a priority review for risks of human cancer from certain exposures to DCH and will conduct a regulatory investigation of the chemical in consultation with other federal agencies (50 FR 42037). The Food and Drug Administration (FDA) regulates the use of chemicals in foods, drugs, and cosmetics. The FDA proposed a ban on the use of DCM as an ingredient in aerosol cosmetic products (primarily hair sprays), based on recent studies which have shown that inhalation of DCM causes cancer in laboratory animals. The FDA maximum permitted residue level of DCM in decaffeinated coffee is 10 ppm. Other FDA regulations applicable to DCM include its use in hops extraction, adhesives, polycarbonate resins, and dilutents in color additive mixtures [as related to food (50 FR 51551)]. The EPA Office of Pesticide Products (OPP) has exempted DCM from the requirement of a tolerance for residues when used as a fumigant after harvest for several grains and citrus fruits. 9.2.2 Advisory Guidance Advisory guidance levels are environmental concentrations recommended by either regulatory agencies or other organizations that are protective of human health or aquatic life. While not enforceable, these levels may be used as the basis for enforceable standards. Advisory guidance for DCM is summarized in Table 9.1 and includes the following: occupational exposure levels, ambient water quality criteria, health advisory levels for drinking water, and suggested-no-adverse- response levels (SNARLs) calculated by the National Academy of Sciences (NAS). The American Conference of Governmental Industrial Hygienists (ACGIH) recently proposed a provisional recommendation to reduce the threshold limit value (TLV) TWA for DCM from 100 to 50 ppm and to eliminate the short-term exposure limit (STEL) in order to provide a wider margin of safety in preventing liver injury. This level should also provide protection against the possible weak carcinogenic effect of DCM that has been demonstrated in laboratory rats and mice. The NIOSH recently changed the Recommended Exposure Limit (REL) for DCM from a TWA of 75 ppm to the lowest feasible limit, based on the assessment of DCM as a potential occupational carcinogen. The NIOSH Immediately Dangerous to Life or Health (IDLH) level for DCM is ------- Regulatory and Advisory Scacus 89 5,000 ppm. This level represents a maximum concentration from which one could escape within 30 min without any escape- impairing symptoms or irreversible health effects. The ODW prepared health advisories (HAs) for numerous drinking water contaminants. The HAs describe concentrations of contaminants in drinking water at which noncarcinogenic effects would not be anticipated to occur and would include a margin of safety to protect sensitive members of the population. The HAs are calculated for One-day, Ten-day, Longer-term, and Lifetime exposures. The One-day HA and Ten-day HA are calculated for exposure of children; for DCM, these values are 13.3 and 1.5 mg/L, respectively. Adequate data for calculating a Longer-term HA were not available. Methylene chloride has been classified by EPA as a B2 carcinogen; therefore, a lifetime HA value is not recommended. However, a DUEL of 1.75 mg/L has been recommended. The NAS calculated One -day and Seven -day SNARLs for DCM. The NAS (1980) listed the One-day SNARL as 35 mg/L and the Seven-day SNARL as 5 mg/L. Subsequent evaluation of these data indicated that values should be 45.5 mg/L and 6.5 mg/L, respectively (EPA 1985g) . Because of the lack of suitable data, a chronic SNARL was not calculated. 9.2.3 Data Analysis 9.2.3.1 Reference doses A reference dose (RfD) has been calculated for DCM. It was based on a 2-year rat study by NCA (1982). In this study, four groups of animals (85 rats/sex/group) were given DCM in drinking water at target doses of 0, 5, 50, 125, and 250 mg/kg/day. Hepatic histological alterations that were detected in the 50- to 250-mg/kg/day dose groups (both sexes) included an increased incidence of foci and areas of cellular alteration. Fatty liver changes were detected in the 125- and 250-mg/kg/day groups at 78 and 104 weeks of treatment. An RfD can be determined using the following general formula: RfD - ' the **" ls the modifying factor . Based on findings of the 2-year rat study, an RfD was calculated (not requiring the use of a modifying factor) as follows: where RfD _ (5 -y-*r—ri . o.05 mgAg/day (100) , 5 mg/kg/day - NOAEL 100 - uncertainty factor appropriate for use with NOAEL from animal study. ------- 90 Section 9 9.2.3.2 Carcinogenic potency In evaluating the potential carcinogenicity of DCM, the EPA Carcinogen Assessment Group (GAG) calculated a human potency estimate by fitting liver and lung tumor data from female B6C3F1 mice in the NTP inhalation study with the linearized multistage model and performing an interspecies extrapolation to humans based on a surface area correction (EPA L985b). The potency factor, q.*, which represents a 95% upper confidence limit of the extra lifetime human risk, is 1.4 x 10'* (mg/kg/day)*^. The unit risk estimate for inhalation exposures is 4.1 x 10'6 (/ig/m3)'1. The EPA (1987a,b) is considering lowering its previous risk estimate (EPA 198Sb) to 4.7 x 10*7 (^/n3)'1 on the basis of pharmacokinetics data reported by Andersen et al (1987). In the risk assessment published in 1985 (EPA 1985b), exposure levels corresponding to excess lifetime human cancer risks of 10'*, 10'5, and 10'6 are 200, 20, and 2 pg/m3 (0.057, 0.0057, and 0.00057 ppm), respectively. The revision based on pharmacokinetics data would raise these exposures by about an order of magnitude. Both risk assessments (EPA 1985b, 1987a,b) are based on the linearized multistage model. Risk estimates made with this model should be regarded as conservative, representing a plausible upper limit for the risk. The true risk is not likely to be higher than the estimate, but it may be lower. To provide a comparison with the linearized multistage model risk estimate, CAG (EPA, 1985b) applied the probit, Ueibull, and time-to-tumor models to the mouse data. The time-to-tumor model estimated risks that were similar to those estimated by the linearized multistage model. For combined lung and liver tumors in female mice, the background-additive implementation of both the probit and Veibull models were in good agreement with the multistage model. However, the background-independent implementations of the probit and Veibull models lead to much lower risk estimates. 9.3 STATE (Regulations and advisory guidance from the states were still being compiled at the time of printing.) 9.3.1 Regulations Methylene chloride is not specified in any state water quality standards. However, this compound is included in the category "toxic substances" in the water quality standards of most states in narrative form. Specific narrative standards protect the use of surface waters for public water supply and contact recreation. 9.3.2 Advisory Guidance No information on state advisory guidance was available. ------- 91 10. REFERENCES ACGIH (American Conference of Government Industrial Hygienists). 1986. Documentation of the Threshold Limit Values and Biological Exposure Indices. 5th ed. Cincinnati, OH. Altshuller AP. 1980. Lifetimes of organic molecules in the troposphere and lower stratosphere. Adv Environ Sci Technol 10:181-219. Anders MW, Kubic VL, Ahmed AE. 1977. Metabolism of halogenated methanes and macromolecular binding. J Environ Pathol Toxicol 1:117-124. Anders MW, Sunram JM. 1982. Transplacental passage of dichloromethane and carbon monoxide. Toxicol Lett 12(4):231-234. Andersen ME, Clewell HJ, Gargas ML, Smith FA, Reitz RH. 1987. Physiologically based pharmacokinetics and the risk assessment process for methylene chloride. Toxicol Appl Pharmacol 87:185-205. Andrae U, Wolff T. 1983. Oichloromethane is not genotoxic in isolated rat hepatocites. Arch Toxicol 52:287-290. Angelo MJ, Pritchard AB. 1984. Simulations of methylene chloride pharmacokinetics using a physiologically based model. Reg Toxicol Pharmacol 4:329-339. * Angelo MJ, Pritchard AB, Hawkins OR, Waller AR, Roberts A. 1986a. The pharmacokinetics of dichloromethane. II. Disposition in Fischer 344 rats following intravenous and oral administration. Food Chem Toxicol 24(9):975-980. * Angelo MJ, Pritchard AB, Hawkins DR, Waller AR, Roberts A. 1986b. The pharmacokinetics of dichloromethane. I. Disposition in B6C3F1 mice following intravenous and oral administration. Food Chem Toxicol 24(9):965-974. APHA (American Public Health Association). 1977. Methods of Air Sampling and Analysis. 2nd ed. Washington, DC: American Public Health Association, pp. 894-902. Key studies. ------- 92 Section 10 APHA (American Public Health Association). 1985. Standard Methods for the Examination of Water and Wastewater. 16th ed. Washington, DC: American Public Health Association. Astrand I, Ovrum P, Carlsson A. 1975. Exposure to methylene chloride. I. Its concentration in alveolar air and blood during rest and exercise and its metabolism. Scand J Work Environ Health 1:78-94. Aviado DM, Belej MA. 1974. Toxicity of aerosol propellants on the respiratory and circulatory systems. I. Cardiac arrythmia in the mouse. Toxicology 2:31-42. Aviado DM, Zakhari S, Watanabe T. 1977. Methylene chloride. In: Goldberg L, ed. Non-Fluorinated Propellants and Solvents for Aerosols, Cleveland, OH: CRC Press, pp. 19-45. Ballantyne B, Guzzard MF, Swanson DW. 1976. Ophthalmic toxicology of dichloromethane. Toxicology 6:173-187 Balmer FM, Smith FA, Leach LJ, Yuile, CL. 1976. Effects in the liver of methylene chloride inhaled along and with ethyl alcohol. Am Ind Hyg Assoc J 37:345-352. Barnes D et al. 1987. Reference dose (RfD): Description and use in health risk assessments. Appendix A in Integrated Risk Information System Supportive Documentation. Vol. 1. Office of Health and Environmental Assessment, Environmental Protection Agency, Washington, DC. EPA/600/8-86/032a., Bellar TA, Lichtenberg JJ, Kroner RC. 1974. The occurrence of organohalides in chlorinated drinking waters. J Am Water Works Assoc 703-706. Berger M, Fodor GG. 1968. CNA disorders under the influence of air mixtures containing dichloromethane. Zentrabl Bakteriol 215:417. Bonventre J, Brennan D, Juson D, Henderson A, Bustos ML. 1977. Two deaths following accidental inhalation of dichloromethane and 1,1,1-trichloroethane. J Anal Toxicol 1:158-160. Bornmann G, Loeser A. 1967. Zur Frage einer Chronisch - toxischen Wirkung von dichloromethan. z. Lebensm-unters. Forsch 136:14-18 (cited in EPA 1985c). Brodzinaki R, Singh HB. 1983. Volatile Organic Chemicals in the Atmosphere: An Assessment of Available Data. Environmental Protection Agency. EPA-600/S 3-83-027. Brunner WD, Staub D, Leisinger T. 1980. Bacterial degradation of dichloromethane. Appl Environ Microbiol 40:950-958. ------- References 93 Burek JD, Nitschke KD, Bell TJ, et al. 1980. Methylene Chloride: A Two-Year Inhalation Toxiclty and Oncogenicity Study in Rats and Hamsters. Toxicology Research Laboratory, Health and Environmental Sciences, Dow Chemical Company, Midland, MI. Burek JD, Nitschke KD, Bell TJ, et al. 1984. Methylene chloride: A two-year inhalation toxicity and oncogenicity study in rats and hamsters. Fundam Appl Toxicol 4(1):30-47. Butler R. Solomon IJ, Snelson A. 1978. Rate constants for the reaction of OH with halocarbons in the presence of 02 + N2. J Air Pollut Control Assoc 28:1131-1133. Carlsson A, Hultengrene M. 1975. Exposure to methylene chloride. III. Metabolism of 14C-labelled methylene chloride in rat. Scand J Work Environ Health 1:104-108. * CEFIC (European Center of Chemical Manufacturer's Federation). 1986a In: Green T, Nash JA, Mainwaring G, eds. Methylene Chloride: In Vitro Metabolism in Rat, Mouse, and Hamster Liver and Lung Fractions and in Human Liver Fractions. ICI Central Toxicology Laboratory, Report CTL/R/879, September 22. CEFIC (European Center of Chemical Manufacturer's Federation). 1986b. In: Green T, Provan WM, Collinge DC, Guest AE, eds. Methylene Chloride Interaction with the Rat and Mouse Liver and Lung DNA In Vivo. ICI Central Toxicology Laboratory, Report CTL/R/851, January 22. CEFIC (European Center of Chemical Manufacturer's Federation). 1986c. In: Truman RW, Ashby J, eds. Methylene Chloride: In Vivo and In Vitro Unscheduled DNA Synthesis Studies in the Mouse and the Rat. ICI Central Toxicology Laboratory, Report CTL/P 1444, January 21. * CEFIC (European Center of Chemical Manufacturer's Federation). 1986d In: Lefevre PA, Ashby J, eds. Methylene Chloride: Induction of S-Phase Hepatocytes in the Mouse After In Vivo Exposure. ICI Central Toxicology Laboratory, Report CTL/R/885, September 22. CEFIC (European Center of Chemical Manufacturer's Federation). 1986e In: Sheldon T, Richardson CR, Hamilton K, Randell V, Hart D, Hollis K, eds. Methylene Chloride: An Evaluation in the Mouse Micronucleus Test. ICI Central Toxicology Laboratory, Report CTL/P/1603, September 19. Cherry N, Venable H, Waldron H. 1983. The acute behavioral effects of solvent exposure. J Soc Occup Med 33:13-18. Clevell HJ, Andersen ME. 1985. Risk assessment extrapolations and physiological modeling. Toxicol Ind Health 1:111-131. ------- 94 Section 10 Coleman WE, Llngg RD, Melton RG, Kopfler FC. 1976. The occurrence of volatile organics in five drinking water supplies using gas chromatography/mass spectrometry. Chap. 21. In: Keith LH, ed. Identification and Analysis of Organic Pollutants in Water. Ann Arbor, HI: Ann Arbor Science. Cox RA, Denwent RC, Eggleton AEJ, Lovelock JE. 1976. Photochemical oxidation of halocarbons in the troposphere. Atmos Environ 10:305-308. CPSC (Consumer Product Safety Commission). 1987. Labeling of certain household products containing methylene chloride; statement of interpretation and enforcement policy. Fed Regist 52(7):34698. Cronn DR. Robinson E. 1979. Determination of trace- gases in Learjet and U-2 whole-air samples collected during the intertropical convergence zone study. In: Poppoff IG, Page WA, Margozzi AP, eds. 1977 Intertropical Convergence Zone Experiment. NASA TMX78577. Cronn DR, Rasmussen RA, Robinson E. 1977. Report for Phase II. Measurement of Tropospheric Halocarbons by Gas Chromatography/Mass Spectrometry. Prepared for the Environmental Protection Agency. Crutzen PJ, Fishman J. 1977. Average concentrations of OH- in the troposphere and the budgets of CH4, CO, H2, and CH3CC13. Geophys Res Lett 4:321-324. Davis DD, Machado G, Conaway B, Oh Y, Watson R. 1976. A temperature- dependent kinetics study of the reaction of OH with CH3C1, CH2C12, CHC13, and CH3. Br J Chem Phys 65(4):1268-1274. Davis EM et al. 1981. Water Res 15:1125-1127. Dewalle FB, Chian ESK. 1978. Proc Ind Waste Conf 32:908-919. Dilling WL, Tefertiller NB, Kallos GJ. 1975. Evaporation rates of methylene chloride, chloroform, 1,1,1-trichloroethane, trichloroethylene, tetrachloroethylene, and other chlorinated compounds in dilute aqueous solutions. Environ Sci Techno1 9(9):833-838. DiVincenzo CD, Yanno FJ, Astill BD. 1971. The gas chromatographic analysis of methylene chloride in breath, blood, and urine. Am Indus Hyg Assoc J 32:387-391. DiVincenzo CD, Yanno FJ, Astill BD. 1972. Human and canine exposure to methylene chloride vapor. Am Ind Hyg Assoc J 33:125-135. DiVincenzo GD, Kaplan CJ. 1981. Uptake, metabolism, and elimination of methylene chloride vapor by humans. Toxicol Appl Pharmacol 59:130-140 Dowty BJ, Carlisle DR, Laseter JL. 1975. New Orleans drinking water sources tested by gas chromatography/mass spectrometry. Environ Sci Technol 9(8):762-765. ------- References 95 Dyksen JE, Hess AF III. 1982. J Am Water Works Assoc 1982:394-403. Elovaara E, Hemminki K, Vainio H. 1979. Effects of methylene chloride. trichloroethane, trichloroethylene, tetrachloroethylene, and toluene on the development of chick embryos. Toxicology 12:111-119. Engstrom J, Bjurstrom R. 1977. Exposure to methylene chloride: Content in subcutaneous adipose tissue. Scand J Work Environ Health 3:215-224. EPA (Environmental Protection Agency). 1975a. Preliminary Assessment of Suspected Carcinogens in Drinking Water. Report to Congress. EPA- 560/14-75-005. PB 260961. EPA (Environmental Protection Agency). 1975b. Region V Joint Federal/State Survey of Organics and Inorganics in Selected Drinking Water Supplies. EPA (Environmental Protection Agency). 1980. Ambient Water Quality Criteria for Halomethanes. Environmental Criteria and Assessment Office, Office of Water Regulations and Standards, Washington, DC. EPA-440/5-80. EPA (Environmental Protection Agency). 1981a. Treatability Manual. EPA-600/2-82-001A, pp. 1.12.2-1 - 1.12.2-4. EPA (Environmental Protection Agency). 1981b. Environmental Risk Assessment of Dichloromethane. Draft report. Office of Toxic Substances. EPA (Environmental Protection Agency). 1983. Methods for Chemical Analysis of Water and Wastes. Office of Research and Development, Environmental Monitoring and Support Laboratory, Cincinnati, OH. EPA-600/4-79-020. EPA (Environmental Protection Agency). 1985a. Health Assessment Document for Dichloromethane. Final report. Office of Health and Environmental Assessment, Washington, DC. EPA/600/8-82/004F. EPA (Environmental Protection Agency). 1985b. Addendum to the Health Assessment Document for Dichloromethane (Methylene Chloride). Updated Carcinogen Assessment of Dichloromethane (Methylene Chloride). Final draft. Office of Research and Development, Washington, DC. EPA 600/8- 82-0044FF. EPA (Environmental Protection Agency). 1985c. Criteria Document on Dichloromethane. Final draft. Office of Drinking Water, Washington, DC. EPA (Environmental Protection Agency). 1985d. Health Effects Assessment for Methylene Chloride. Office of Health and Environmental Assessment, Washington, DC. Cincinnati, OH: Office of Emergency and Remedial Response. ------- 96 Section 10 EPA (Environmental Protection Agency). 1985e. Environmental Profiles and Hazard Indices for Constituents of Municipal Sludge: Methylene Chloride. Office of Water Regulations and Standards, Criteria and Standards Division. EPA (Environmental Protection Agency). 1985f. Summary of Environmental Profiles and Hazard Indices for Constituents of Municipal Sludge: Methods and Results. Office of Water Regulations and Standards, Criteria and Standards Division. EPA (Environmental Protection Agency). 1985g. Health Advisory for Dichloromethane. Draft. Office of Drinking Water, Washington, DC. EPA (Environmental Protection Agency). 1985h. Analysis of the Applicability of TSCA Section 4(f) to Methylene Chloride. Prepared by J. Hopkins. EPA (Environmental Protection Agency). 1986a. Superfund Public Health Evaluation Manual. Office of Emergency and Remedial Response, Washington, DC. EPA 540/1-86-060. EPA (Environmental Protection Agency). 1986b. Test Methods for Evaluating Solid Waste. 3rd ed. Office of Solid Waste and Emergency Response, Washington, DC. SW-846. EPA (Environmental Protection Agency). 1986c. Guidelines for carcinogen risk assessment. Fed Regist 51(195):33992-34003. * EPA (Environmental Protection Agency). 1987a. Technical Analysis of New Methods and Data Regarding Dlchloromethane Hazard Assessments. Draft document. * EPA (Environmental Protection Agency). 1987b. Update to Health Assessment Document and Addendum for Dichloromethane (Methylene Chloride): Pharmacokinetics, Mechanism of Action, and Epidemiology. Draft report. EPA 600/8-87/1030A. Ewing BB, Chi an ESK. Cook JC, Evans CA, Hopke PK, Perkins EG. 1977. Monitoring to Detect Previously Unrecognized Pollutants in Surface Waters. Environmental Protection Agency. EPA-560/6-77-015. Flury Fanil Zernik F. 1931. Harmful Gases, Vapors, Fog, Smoke, and Dust. Berlin: Julius Springer, pp. 311-312. Fodor GG, Winneke G. 1971. Nervous system disturbances in men and animals experimentally exposed to industrial solvent vapors in England. Proceedings of the 2nd International Clean Air Congress. New York: Academic Press. Fodor GG, Prajsnar D, Schlipkoter HW. 1973. Endogenous cofornation by incorporated halogenated hydrocarbons of the methane series. Staubreinhalt Luft 33:260-261. ------- References 97 Fodor GG, Roscovana G. 1976. Increased blood-CO content in humans and animals by incorporated halogenated hydrocarbons. Zentrabl Bukteriol (Orig B) 162:34 (German) (cited in EPA 1985a). Forster HV, Graff S, Hake CL, Soto R, Stewart RD. 1974. Pulmonary- Hematologic Studies on Humans During Exposure to Methylene Chloride. NIOSH-MCOW-ENVM-MC-74-4. Friedlander BR, Hearne FT. Hall S. 1978. Epidemiologic investigation of employees chronically exposed to methylene chloride--mortality analysis J Occup Hed 20(10):657-666. Gargas ML, Clewell HJ, Andersen ME. 1986. Metabolism of inhaled dihalomethanes in vivo: Differentiation of kinetic constants for two independent pathways. Toxicol Appl Pharmacol 82:211-223. Gocke E, King M-T, Eckhardt K, Wild D. 1981. Mutagenicity of cosmetic ingredients licensed by European Communities. Mutat Res 90:91-109. Gradiski D, Bonnet G, Raoult G, Magadur JL. 1978. Compared acute pulmonary toxicity of the main chlorinated aliphatic solvents (French). Arch Mai Prof Med Trav Secus Soc 39:249. Grimsrud EP, Rasmussen RA. 1975. Survey and analysis of halocarbons in the atmosphere by gas chromatography-mass spectrometry. Atmos Environ 9:1014-1017. Hardin BD, Manson JM. 1980. Absence of dichloromethane teratogenicity with inhalation exposure to rats. Toxicol Appl Pharmacol 52(1):22. Harkov R, Giante SJ Jr., Bozelli JW, LaRegina JE. 1985. Monitoring volatile organic compounds at hazardous and sanitary landfills in New Jersey. J Environ Sci Health A20(5):491-501. * Haun CC, Vernot EH, Darmer KI, Diamond SS. 1972. Continuous animal exposure to low levels of dichloromethane. In: Proceedings of the Third Annual Conference on Environmental Toxicology. Wright-Patterson Air Force Base, OH: Aerospace Medical Research Laboratory. AMRL-TR-72-130, pp. 199-208. Hearne FT, Grose F, Pifer JW. Friedlander BR, Raleigh RL. 1987. Methylene chloride mortality study: Dose-response characterization and animal model comparison. J Occup Med 29:217-228. Helz GR, Hus RY. 1978. Limnol Oceanogr 23:858-869. Heppel LA, Neal PA. 1944. Toxicology to dichloromethane (methylene chloride). II. Its effects upon running activity in the male rat. J Indust Hyg Toxicol 26:17-21. Heppel LA, Neal PA, Perrin ML, On VT. 1944. Toxicology of dichloromethane (methylene chloride). I. Studies on effects of daily inhalation. J Indust Hyg Toxicol 26 8-16. ------- 98 Section 10 Hlatt MH. 1981. Analysis of fish and sediment for volatile priority pollutants. Anal Chem 53:1541-1543. HSDB (Hazardous Substances Data Bank). 1987. Record for dichloromethane Computer printout. National Library of Medicine, April 9, 1987. HSIA (Halogenated Solvents Industry Alliance). 1988. Public comments on scientific and technical issues on the Draft Toxicological Profile for Methylene Chloride. Submitted to Agency for Toxic Substances and Disease Registry (ATSDR) March 8. IARC (International Agency for Research on Cancer). 1986. Some halogenated hydrocarbons and pesticide exposures. IARC Monograph on the Evaluation of the Carcinogenic Risk of Chemicals to Humans. 41:43-85. ICAIR. 1985. Chemical Profile: Methylene Chloride. ICAIR Life Systems Cleveland, OH. TR-894-30. Illing HPA, Shillaker RO. 1985. Toxicity Review 12. Dichloromethane (Methylene Chloride). Health and Safety Executive. Her Majesty's Stationary Office, London, England. Jongen WMF, Lehman PHM, Kottenhagen MJ, Alink GM, Berends F. Koeman JH. 1981. Mutagenicity testing of dichloromethane in short-term mammalian test systems. Mutat Res 81:203-213. Kimura ET, Ebert DM, Dodge PW. 1971. Acute toxicity and limits of solvent residue for sixteen organic solvents. Toxicol Appl Pharmacol 19:699-704. Klecka GM. 1982. Fate and effects of methylene chloride in activated sludge. Appl Environ Microbiol 44:701-707. Konasewich D et al. 1978. Status Report on Organic and Heavy Metal Contaminants in the Lakes Erie, Michigan, Huron, and Superior Basins. Great Lake Water Quality Board, p. 373. Kuzelova MR, Vlasak. 1966. The effect of methylene dichloride on the health of workers in production of film foils and investigation of formic acid as the methylene dichloride metabolite (Czech). Pracov Lek 18:167-170 (cited in NIOSH 1976). LaPat-Polasko LT, McCarty PL, Zehnder AJB. 1984. Secondary substrate utilization of methylene chloride by an isolated strain of Pseudononas Appl Environ Microbiol 47(4):825-830. Latham S, Potvln M. 1976. Microdetermination of dichloromethane in blood with a syringeless gas chromatographic injection system. Chemosphere 6:403-411. MacEwen JD, Vernot EH, Haun CC. 1972. Continuous Animal Exposure to Dichloromethane. Wright-Patterson Air Force Base, OH: Aerospace Medical Research Laboratory. AMRL-TR-72-28. ------- References 99 Maksimov GG, Mamleeva NK, Malyarova LK. 1977. Distribution, accumulation, and excretion of methylene chloride following its entry through the skin (Russian) . Kozhn Put Postupleniya Prom Yadov v Organizm. i Ego Prof ila-ktika: 17-21 (cited in EPA 1985c) . McCarroll NE, Cortina TA, Zieo HJ , Farrow MG. 1983. Evaluation of methylene chloride and vinylidene chloride in mutational assays. Environ Mutagenesis 5:426-427. McConnell FE, Solleveld HA, Swenberg JA, Boorman GA. 1986. Guidelines for combining neoplasms for evaluation of rodent carcinogenesis studies. J Natl Cancer Inst 76:283-289. McDougal JN, Jepson GV, Clewell HJ , MacNaughton MG, Andersen ME. 1986. A physiological pharmacokinetic model for dermal absorption of vapors in the rat. Toxicol Appl Pharmacol 85:286-294. McKenna MJ, Saunders JH, Boeckler WH, Karbowski RJ , Nitschke KD, Chenoweth MB. 1980. The pharmacokinetics of inhaled methylene chloride in human volunteers. Toxicol Appl Pharmacol A59 (abstract). * McKenna MJ, Zempel JA. 1981. The dose -dependent metabolism of methylene chloride following oral administration to rat. Food Cosmet Toxicol 19:73-78. * McKenna MJ, Zempel JA, Braun WH. 1982. The pharmacokinetics of inhaled methylene chloride in rats. Toxicol Appl Pharmacol 65:1-10. Moody DE. 1981. Correlations among changes in hepatic microsomal components after intoxication with alkyl halides and other hepatotoxins . Mol Pharmacol 20:685-693. Morris JB, Smith FA, Carman RH. 1979. Studies on methylene chloride- induced fatty liver. Exp Mol Pathol 30:386-393. NAS (National Academy of Sciences). 1977. Drinking Water and Health. Washington, DC: National Academy Press, pp. 743-745. NAS (National Academy of Sciences). 1978. Non- Fluor ina ted Halomethanes in the Environment. Panel on Low-Molecular-Weight Halogenated Hydrocarbons. Coordinating Committee for Scientific and Technical Assessments of Environmental Pollutants. NAS (National Academy of Sciences). 1980. Drinking Water and Health. Vol. 3. Washington, DC: National Academy Press. * NCA [National Coffee Association (prepared by Hazelton Laboratories American, Inc.)]. 1983. 24-Month Oncogenicity Study of Methylene Chloride in Mice. Vienna, VA: Unpublished (reported in Serota et al. 1984) . ------- 100 Section 10 * NCA [National Coffee Association (prepared by Hazelton Laboratories American, Inc.)]. 1982. 24-Month Chronic Toxicity and Oncogenicity Study of Methylene Chloride in Rats. Final report. Vols. I-IV. Vienna, VA: Unpublished. 2112-101. NFPA (National Fire Protection Association). 1978. Fire Protection Guide for Hazardous Materials. NIOSH (National Institute for Occupational Safety and Health). 1976. Criteria for a Recommended Standard...Occupational Exposure to Methylene Chloride. Cincinnati. OH. U.S. DHEW Publ. 76-138. NIOSH (National Institute for Occupational Safety and Health). 1984. NIOSH Manual of Analytical Methods. Vol. 2. 3rd ed. Cincinnati, OH: Department of Health and Human Services. NIOSH (National Institute for Occupational Safety and Health). 198S. NIOSH Pocket Guide to Chemical Hazards. Washington, DC: Department of Health and Human Services. NIOSH (National Institute for Occupational Safety and Health). 1986. Current Intelligence Bulletin 46 - Methylene Chloride. Cincinnati, OH: Department of Health and Human Services. Nitschke KD, Burek JD, Bell TJ, Kociba RJ, Rampy LW, McKenna MJ. 1988. Methylene chloride: A 2-year inhalation toxicity and oncogenicity study in rats. Fundam Appl Toxicol 11:48-59. * Nitschke K, Eisenbrandt DL, Lomax LC. 1985. Methylene chloride: Two- generation inhalation reproduction study in Fischer 344 rat. Halogenated Solvents Industry Alliance, Unpublished (currently being peer reviewed, October/November 1987). Nitschke KD, Eisenbrandt DL, Lomax LG, Rao KS. 1988. Methylene chloride. Two-generation inhalation reproductive study in rats. Fundam Appl Toxicol 11:60-67. * Nitschke KD, Burek JD, Bell TJ, Rampy LW. McKenna MG. 1982. Methylene Chloride: A Two-Year Inhalation Toxicity and Oncogenicity Study. Toxicology Research Laboratory, Health and Environmental Sciences, Dow Chemical. Final report. Norpoth K, Witting U, Springoram M, Wittig C. 1974. Induction of microsomal enzymes in the rat liver by inhalation of hydrocarbon solvents. InC Arch Arbeitmed 33(4):315-321 (cited in EPA 1985). * NTP (National Toxicology Program). 1986. Toxicology and Carcinogenesis Studies of Dichloromethane (Methylene Chloride) (CAS 75-09-2) in F344/N Rats and B6C3F1 Mice (Inhalation Studies). Tech Rep Ser 306. ORVWSC (Ohio River Valley Water Sanitary Commission). 1982. Assessment of Wa«:er Quality Conditions. Ohio River Mainstream. 1981-1982. Table 13 ------- References 101 OSHA (Occupational Safety and Health Administration). 1979. General Industry Standards. (OSHA) 2206, Revised January 1978. Department of Labor, Washington, DC. OSHA (Occupational Safety and Health Administration). 1986. Occupational exposure to methylene chloride. Fed Regist 51:42257. Ott MG, Skory LK, Holder BB. Bronson JM, Williams PR. 1983a. Health evaluation of employees occupationally exposed to methylene chloride - mortality. Scand J Work Environ Health 9(Suppl 1):8-16. Ott MG, Skory LK, Holder BB, Bronson JM, William PR. 1983b. .Health evaluation of employees occupationally exposed to methylene chloride Twenty-four hour electrocardiographic monitoring. Scand J Work Environ Health 9:26-30. * Ott MG, Skory LK, Holder BB, Bronson JM, William PR. 1983c. Health evaluation of employees occupationally exposed to methylene chloride. Metabolism data and oxygen half saturation pressure. Scand J Work Environ Health 9:31-38. Page BD, Charbonneau CF. 1977. Gas chromatographic determination of residual methylene chloride and trichloroethylene in decaffeinated instant and ground coffee with electrolytic conductivity and electron capture detection. J Assoc Off Anal Chem 60:710-715. Page BD, Charbonneau CF. 1984. Headspace gas chromatographic determination of methylene chloride in decaffeinated tea and coffee, with electrolytic detection. J Assoc Off Anal Chem 67:757-761. Page BD, Kennedy BPC. 1975. Determination of methylene chloride. ethylene dichloride, and trichloroethylene as solvent residues in spice oleoresins, using vacuum distillation and electron capture gas chromatography. J Assoc Off Anal Chem 58:1062-1068. Pearson CR, McConnell G. 1975. Chlorinated Cl and C2 hydrocarbons in the marine environment. Proc R Soc London B 189:305-332. Pellizzari ED. 1974. Electron capture detection in gas chromatography. J Chromatogr 98:323-361. Pellizzari ED. 1977. Analysis of Organic Air Pollutants by Gas Chromatography and Mass Spectroscopy. Environmental Protection Agency EPA-600/2-77-100. Pellizzari ED. 1978a. Measurement of Carcinogenic Vapors in Ambient Atmospheres. Environmental Protection Agency. EPA-600/7-78-062. Pellizzari ED. 1978b. Quantification of Chlorinated Hydrocarbons in Previously Collected Air Samples. Environmental Protection Agency. EPA-450/3-78-112. ------- 102 Section 10 Pellizzari ED, Bunch JE. 1979. Ambient Air Carcinogenic Vapors: Improved Sampling and Analytical Techniques and Field Studies. Environmental Protection Agency. EPA-600/2-79-081. Pellizzari ED, Erickson MD, Zweidinger RA. 1979. Formulation of a Preliminary Assessment of Halogenated Organic Compounds in Man and Environmental Media. Environmental Protection Agency. EPA-560/13-179-006 Perocco P, Prodi G. 1981. DNA damage by halokanes in human lymphocytes cultured in vitro. Cancer Lett 13:213-218. Peterson JE. 1978. Modeling the uptake, metabolism, and excretion of dichloromethane by man. Am Ind Hyg Assoc J 39:41-47. Ramsey JC, Andersen ME. 1984. A physiologically based description of the inhalation pharmacoklnetics of styrene in rats and humans. Toxicol Appl Pharmacol 73:159-175. Ramussen RA, Harsch DE, Sweany PH, Krasnec JP, Cronn DR. 1979. Determination of atmospheric halocarbons by a temperature programmed gas chromatographic freezeout concentration method. J Air Pollut Control Assoc 27:579. Ratney RS, Wegman DH, Elkins HB. 1974. In vivo conversion of methylene chloride to carbon monoxide. Arch Environ Health 28:223-226. Riley EC, Fasset DW, Sutton WL. 1966. Methylene chloride vapor in expired air of human subjects. Am Ind Hyg Assoc J 27:341-348. Rittman BE, McCarty PL. 1980. Utilization of dichloromethane by suspended and fixed-film bacteria. Appl Environ Microbiol 39:1225-1226 Rodkey FL, Collison HA. 1977. Effect of dihalogenated methanes on the in vivo production of carbon monoxide and methane by rats. Toxicol Appl Pharmacol 40:39-47. Robinson E. 1978. Analysis of Halocarbons in Antarctica. Report 78/13-42 prepared for the National Science Foundation. Roth RP, Drew RT, Lo RJ, Fouts JR. 1975. Dichloromethane inhalation, carboxyhemoglobin concentrations, and drug metabolizing enzymes in rabbits. Toxicol Appl Pharmacol 33:427-437. Ruth JH. 1986. Odor thresholds and initatlon levels of several chemical substances. A review. Am Ind Hyg Assoc J 47:A142-A151. Savolainen H, Pfaffli P, Tengen M, Vainio H. 1977. Biochemical and behavioral effects of inhalation exposure to tetrachloroethylene and dichloromethane. J Neuropath Exp Neurol 36:941-949. ------- References 103 Schvetz BA, Leong BJ, Gehrlng PJ. 1975. The effect of maternally inhaled trichloroethylene, perchloroethane, methyl chloroform, and methylene chloride on embryonal and fetal development in mice and rats. Toxicol Appl Pharmacol 32:84-96. Serota D, Ulland B. Carlborg F. 1984. Hazleton Chronic Oral Study in Mice. Food Solvents Workshop No. 1. Methylene Chloride. March 8-9, Bethesda, MD. In: Anderson ME, Clewell HJ, Gargas ML, Smith FA, Reitz RH. 1987. Physiologically based pharmacokinetics and the risk assessment process for methylene chloride. Toxicol Appl Pharmacol 87:185-205. Singh HB. 1977. Atmospheric halocarbons. Evidence in favor of reduced average hydroxyl radical concentrations in the troposphere. Geophys Res Lett 4(3):101-104. Singh HB et al. 1982. Environ Sci Technol 16:872-880. Singh HB, Salas LJ, Shigeishi H, Smith AJ, Scribner E, Cavanagh LA. 1979. Atmospheric Distributions, Sources, and Sinks of Selected Halocarbons, Hydrocarbons, SF6, and N20. Final report submitted to the Environmental Protection Agency by SRI International, Menlo Park, CA. EPA-600/3-79-107. Singh HB, Salas LJ, Smith AJ, Shigeishi. 1981. Measurements of some potentially hazardous organic chemicals in urban environments. Atmos Environ 15:601-612. Singh HB, Salas LJ, Stiles RE. 1983. Selected man-made halogenated chemicals in the air and oceanic environment. J Geophys Res 88:3675-3683. Singh HB, Salas LJ, Stiles RE, Shigeishi H. 1980. Atmospheric Measurements of Selected Hazardous Organic Chemicals. Second Year Interim Report. Prepared for the Environmental Protection Agency by SRI International, Menlo Park, CA. Stewart RD, Fischer TN, Hosko HJ, Peterson JE, Bare tea ED, Dodd HC. 1972. Experimental human exposure to methylene chloride. Arch Environ Health 25:342-348. Stewart RD, Hake CL. 1976. Paint remover hazard. J Med Assoc 235(4):398-401. Stover EL, Kincannon DF. 1983. J Water Pollut Control Fed 55:97-109. Svirbely JL, Highman B, Alford WF, Von Oettinger WF. 1947. The toxicity and narcotic action of mono-chloro-mono-bromo-methane with special reference to inorganic and volatile bromide in blood, urine, and brain. J Ind Hyg Toxicol 23:383. Tabak HH et al. 1981. J Water Pollut Control Assoc 53:1503-1518. ------- 104 Section 10 Thilagar AK, Back AM, Kirby PE, et al. 1984a. Evaluation of dlchloromethane in short-tern in vitro genetic toxicity assays. Environ Mutagenesis 6:418-419. Thilagar AK, Kumaroo PV, Clark JJ, Kott A, Back AM, Kirby PE. 1984b. Induction of chromosome damage by dichloromethane in cultured human peripheral lymphocytes, CHO cells, and mouse lymphoma LS178Y cells. Environ Mutagenesis 6:422. Thilagar AK, Kumaroo V. 1983. Induction of chromosome damage by methylene chloride in CHO cells. Mutat Res 116:361-367. Thomas AA, Pinkerton MK, Warden JA. 1972. Effects of low-level dichloromethane exposure on the spontaneous activity of mice. In: Proceedings of the 3rd Annual Conference on Environmental Toxicology. Wright-Patterson Air Force Base, OH: Aerospace Medical Research Laboratory. AMRL-TR72-130, pp. 185-189. Ugazio G, Burdino E, Danni 0, Milillo PA. 1973. Hepatoxicity and lethality of halogenoalkanes. Biochem Soc Trans 1:968-972. Verschueren K. 1977. Handbook of Environmental Data on Organic Chemicals. New York: Van Nostrand Reinhold. Von Oettingen WF, Powell CC, Sharpless NE. Alford WC, Pecora LJ. 1949. Relation Between the Toxic Action of Chlorinated Methanes and Their Chemical and Physicochemical Properties. NIH Bulletin 191. Federal Security Agency, U.S. Public Health Service, National Institutes of Health. Weast RC, ed. 1985. CRC Handbook of Chemistry and Physics. 66th ed. Boca Raton, FL: CRC Press. Weinstein RS, Diamond SS. 1972. Hepatotoxicity of dichloromethane (methylene chloride) with continuous inhalation exposure at a low dose level. In:. Proceedings of the 3rd Annual Conference on Environmental Toxicology. Wright-Patterson Air Force Base, OH: Aerospace Medical Research Laboratory. AMRL-TR-72-130, pp. 209-220. Weinstein RS, Boyd DD, Back KC. 1972. Effects of continuous inhalation of dichloromethane in the mouse--morphologic and functional observations. Toxicol Appl Pharmacol 23:660 (cited in EPA 1985c). Weiss G. 1967. Toxic encephalosis as an occupational hazard with methylene chloride. Zentralbl Arbeitsmed 17:282-285. Welch L. 1987. Reports of clinical disease secondary to methylene chloride exposure--a collection of 141 cases. Unpublished study. Submitted to OPTS/EPA 3/31. ------- References 105 * Wlnneke G. 1974. Behavioral effects of methylene chloride and carbon monoxide as assessed by sensory and psychomotor performance. In: Xintaraa C, Johnson BL, de Groot I, eds. Behavioral Toxicology. Washington, DC: U.S. Government Printing Office, pp. 130-144. Wood PR, Parsons FZ, OeMarco J, et al. 1981. Introductory study of the biodegradation of ethane and ethene compounds. Presented'at the American Water Works Association meeting, June. Yesair DW, Jacques D. Schepis P, Liss RH. 1977. Dose-related pharmacokinetics of 14C methylene chloride in mice. Fed Proc 36:998 (cited in EPA 1985c). ------- 107 11. GLOSSARY Acute Exposure--Exposure to a chemical for a duration of 14 days or less, as specified in the Toxicological Profiles. Bloconcentratlon Factor (BCF)--The quotient of the concentration of a chemical in aquatic organisms at a specific time or during a discrete time period of exposure divided by the concentration in the surrounding water at the same time or during the same time period. Carcinogen--A chemical capable of inducing cancer. Ceiling value (CL)--A concentration of a substance that should not be exceeded, even instantaneously. Chronic Exposure--Exposure to a chemical for 365 days or more, as specified in the Toxicological Profiles. Developmental Toxicity--The occurrence of adverse effects on the developing organism that may result from exposure to a chemical prior co conception (either parent), during prenatal development, or postnatally to the time of sexual maturation. Adverse developmental effects may be detected at any point in the life span of the organism. Embxyotoxlclty and Fetotoxicity--Any toxic effect on the conceptus as a result of prenatal exposure to a chemical; the distinguishing feature between the two terms is the stage of development during which the insult occurred. The terms, as used here, include malformations and variations, altered growth, and in utero death. Frank Effect Level (PEL)--That level of exposure which produces a statistically or biologically significant increase in frequency or severity of unmistakable adverse effects, such as irreversible functional impairment or mortality, in an exposed population when compared with its appropriate control. EPA Health Advisory--An estimate of acceptable drinking water levels for a chemical substance based on health effects information. A health advisory is not a legally enforceable federal standard, but serves as technical guidance to assist federal, state, and local officials. Immediately Dangerous to Life or Health (IDLH)--The maximum environmental concentration of a contaminant from which one could escape within 30 min without any escape-impairing symptoms or irreversible health effects. ------- 108 Sec don 11 Intermediate Exposure--Exposure to a chemical for a duration of 15-364 days, as specified in the Toxicological Profiles. Inmmnologic Toxicity--The occurrence of adverse effects on the immune system that may result from exposure to environmental agents such as chemicals. In vitro--Isolated from the living organism and artificially maintained. as in a test tube. In vivo--Occurring within the living organism. Key Study--An animal or human toxicological study that best illustrates the nature of the adverse effects produced and the doses associated with those effects. Lethal Coneentration(LO) (LCLO)--The lowest concentration of a chemical in air which has been reported to have caused death in humans or animals. Lethal Concentration(SO) (LCso)--A calculated concentration of a chemical in air to which exposure for a specific length of time is expected to cause death in 50% of a defined experimental animal population. Lethal Dose(LO) (LDLO)--The lowest dose of a chemical introduced by a route other than inhalation that is expected to have caused death in humans or animals. Lethal Dose(50) (U>50)--The dose of a chemical which has been calculated to cause death in 50% of a defined experimental animal population. Lowest-Observed-Adverse-Effect Level (LOAEL)--The lowest dose of chemical in a study or group of studies which produces statistically or biologically significant increases in frequency or severity of adverse effects between the exposed population and its appropriate control. Lowest-Observed-Effect Level (LOEL)--The lowest dose of chemical in a study or group of studies which produces statistically or biologically significant increases in frequency or severity of effects between the exposed population and its appropriate control. Malformation*--Permanent structural changes that may adversely affect survival, development, or function. Minimal Risk Laval--An estimate of daily human exposure to a chemical that is likely to be without an appreciable risk of deleterious effects (noncancerous) over a specified duration of exposure. Mutagen--A substance that causes mutations. A mutation is a change in the genetic material in a body cell. Mutations can lead to birth defects, miscarriages, or cancer. ------- Glossary 109 Neurotoxicity--The occurrence of adverse effects on the nervous system following exposure to a chemical. No-Observed-Adverse-Effect Level (NOAEL)--That dose of chemical at which there are no statistically or biologically significant increases in frequency or severity of adverse effects seen between the exposed population and its appropriate control. Effects may be produced at this dose, but they are not considered to be adverse. No-Observed-Effect Level (NOEL)--That dose of chemical at which there are no statistically or biologically significant increases in frequency or severity of effects seen between the exposed population and its appropriate control. Permissible Exposure Limit (PEL)--An allowable exposure level in workplace air averaged over an 8-h shift. q.*--The upper-bound estimate of the low-dose slope of the dose-response curve as determined by the multistage procedure. The q * can be used to calculate an estimate of carcinogenic potency, the incremental excess cancer risk per unit of exposure (usually pg/L for water, mg/kg/day for food, and pg/w? for air). Reference Dose (RfD)--An estimate (with uncertainty spanning perhaps an order of magnitude) of the daily exposure of the human population to a potential hazard that is likely to be without risk of deleterious effects during a lifetime. The RfD is operationally derived from the NOAEL (from animal and human studies) by a consistent application of uncertainty factors that reflect various types of data used to estimate RfDs and an additional modifying factor, which is based on a professional judgment of the entire database on the chemical. The RfDs are not applicable to nonthreshold effects such as cancer. Reportable Quantity (RQ)--The quantity of a hazardous substance that is considered reportable under CERCLA. Reportable quantities are: (1) 1 Ib or greater or (2) for selected substances, an amount established by regulation either under CERCLA or under Sect. 311 of the Clean Water Act. Quantities are measured over a 24-h period. Reproductive ToxicIty--The occurrence of adverse effects on the reproductive system that may result from exposure to a chemical. The toxicity may be directed to the reproductive organs and/or the related endocrine system. The manifestation of such toxicity may be noted as aIterations-in sexual behavior, fertility, pregnancy outcomes, or modifications in other functions that are dependent on the integrity of this system. Short-Term Exposure Limit (STEL)--The maximum concentration to which workers can be exposed for up to IS min continually. No more than four excursions are allowed per day, and there must be at least 60 min between exposure periods. The dally TLV-TWA may not be exceeded. ------- 110 Section 11 Target Organ ToxicIty--This term covers a broad range of adverse effects on target organs or physiological systems (e.g., renal, cardiovascular) extending from those arising through a single limited exposure to those assumed over a lifetime of exposure to a chemical. Teratogen--A chemical that causes structural defects that affect the development of an organism. Threshold Limit Value (TLV)--A concentration of a substance to which most workers can be exposed without adverse effect. The TLV may be expressed as a TWA, as a STEL, or as a CL. Time-velghted Average (TWA)--An allowable exposure concentration averaged over a normal 8-h workday or 40-h workweek. Uncertainty Factor (OF)--A factor used in operationally deriving the RfD from experimental data. UFs are intended to account for (1) the variation in sensitivity among the members of the human population, (2) the uncertainty in extrapolating animal data to the case of humans, (3) the uncertainty in extrapolating from data obtained in a study that is of less than lifetime exposure, and (4) the uncertainty in using LOAEL data rather than NOAEL data. Usually each of these factors is sec equal to 10. ------- Ill APPENDIX: PEER REVIEW A peer review panel was assembled for methylene chloride. The panel consisted of Che following members: Dr. Herbert Cornish (retired). University of Michigan; Dr. Rudolph Jaeger (consultant), New York University Medical Center; and Mr. L. Skory (retired), Dow Chemical Company. These experts collectively have knowledge of methylene chloride's physical and chemical properties, toxicokinetics, key health end points, mechanisms of action, human and animal exposure, and quantification of risk to humans. All reviewers were selected in conformity with the conditions for peer review specified in the Superfund Amendments and Reauthorization Act of 1986, Section 110. A Joint panel of scientists from ATSDR and EPA has reviewed the peer reviewers' comments and determined which comments will be included in the profile. A listing of the peer reviewers' comments not incorporated in the profile, with a brief explanation of the rationale for their exclusion, exists as part of the administrative record for this compound. A list of databases reviewed and a list of unpublished documents cited are also included in the administrative record. The citation of the peer review panel should not be understood to imply their approval of the profile's final content. The responsibility for the content of this profile lies with the Agency for Toxic Substances and Disease Registry. ------- |